1
|
Shubow S, Gunsior M, Rosenberg A, Wang YM, Altepeter T, Guinn D, Rajabiabhari M, Kotarek J, Mould DR, Zhou H, Cheifetz AS, Garces S, Chevalier R, Gavan S, Trusheim MR, Rispens T, Bray K, Partridge MA. Therapeutic Drug Monitoring of Biologics: Current Practice, Challenges and Opportunities - a Workshop Report. AAPS J 2025; 27:62. [PMID: 40087239 DOI: 10.1208/s12248-025-01050-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/23/2025] [Indexed: 03/17/2025] Open
Abstract
Therapeutic drug monitoring (TDM) for dose modification of biologics has the potential to improve patient outcomes. The US Food and Drug Administration (FDA) and the American Association of Pharmaceutical Scientists (AAPS) hosted the first US-based public workshop on TDM of biologics with contributions from a broad array of interested parties including healthcare providers, clinical pharmacologists, test developers, bioanalysis and immunogenicity scientists, health economics and outcomes research (HEOR) experts and regulators. The key insight was that despite a body of evidence to support TDM in certain therapeutic areas, there remain substantial challenges to widespread clinical implementation. There is a lack of consensus regarding the integration of TDM in clinical guidelines, and a lack of consensus on the cost-effectiveness of TDM; both factors contribute to the difficulty that healthcare providers face in obtaining reimbursement for TDM (both coverage of testing itself, and coverage of potential dosing modifications). The HEOR experts outlined alternative routes to obtaining reimbursement and suggested advocating for changes in coverage policies to promote TDM use in the clinic. Reaching alignment across policy makers, patients and advocacy groups, payers, and healthcare providers, on specific treatment settings where TDM will be clearly beneficial, was identified as an important step to advancing TDM implementation for the benefit of patients.
Collapse
Affiliation(s)
- Sophie Shubow
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Yow-Ming Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Tara Altepeter
- Division of Gastroenterology, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Daphne Guinn
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Joseph Kotarek
- Office of Health Technology 7, Office of Product Evaluation and Quality, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Diane R Mould
- Projections Research Inc., Phoenixville, Pennsylvania, USA
| | - Honghui Zhou
- Jazz pharmaceuticals, Philadelphia, Pennsylvania, USA
| | - Adam S Cheifetz
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Rachel Chevalier
- Children's Mercy Kansas City, University of Missouri-Kansas City (UMKC), Kansas City, USA
| | - Sean Gavan
- Manchester Centre for Health Economics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | | | - Theo Rispens
- Amsterdam institute for Immunology and Infectious diseases, Immunology, Amsterdam, Netherlands
| | | | | |
Collapse
|
2
|
Ortiz-Fernández P, Iniesta-Navalón C, Urbieta-Sanz E, Gascón-Cánovas JJ. Assessing early therapeutic drug monitoring of adalimumab as a predictor of treatment efficacy and immunogenicity in rheumatic diseases: "early therapeutic drug monitoring of adalimumab". Clin Rheumatol 2025; 44:1009-1018. [PMID: 39826047 DOI: 10.1007/s10067-025-07307-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/17/2024] [Accepted: 12/29/2024] [Indexed: 01/20/2025]
Abstract
INTRODUCTION Therapeutic drug monitoring (TDM) in inflammatory rheumatic diseases (RMDs) is gaining interest. However, there are unresolved questions about the best practices for implementing TDM effectively in clinical settings. OBJECTIVE The primary objective of this study was to evaluate whether early TDM of adalimumab predicts drug survival at 52 weeks in patients with RMDs. The secondary objective was to identify factors associated with pharmacokinetic failure and treatment discontinuation. METHODS A retrospective cohort study included patients aged ≥ 18 years with RMDs who initiated adalimumab therapy. Early TDM was performed within the first 26 weeks, and adalimumab trough levels (ATL) and anti-drug antibodies were measured. Drug survival was assessed at 52 weeks and defined as the time from adalimumab initiation to discontinuation for any reason. Multivariate analyses were conducted to identify factors influencing outcomes. RESULTS The study included 194 patients, of whom 56.7% exhibited ATL below the therapeutic range during the first 26 weeks. In the multivariate analysis, subtherapeutic concentrations were significantly associated with higher weight (OR = 1.02; p = 0.040) and ankylosing spondylitis diagnosis (OR = 3.68; p < 0.001). At 52 weeks, 43.8% of patients had discontinued adalimumab. Low ATL (< 1 µg/mL) was strongly associated with treatment discontinuation (OR = 7.31; p < 0.001), while concomitant methotrexate reduced this risk (OR = 0.46; p = 0.026). CONCLUSIONS Early TDM of adalimumab predicts drug persistence and underscores its clinical relevance as a proactive tool to guide personalized treatment and reduce the risk of treatment failure. These findings highlight the importance of incorporating TDM into routine practice to optimize therapeutic outcomes. Key Points • Early TDM of adalimumab in rheumatic diseases shows that low drug exposure predicts reduced drug survival at 52 weeks. • Approximately half of the patients exhibit low adalimumab exposure with the standard dose (40 mg every other week). • Body weight and methotrexate use significantly impact adalimumab levels. • Immunogenicity, found in 14.4% of patients with low ADL levels, underscores the need for early ADA detection to prevent non-response and discontinuation.
Collapse
Affiliation(s)
- Patricia Ortiz-Fernández
- Department of Hospital Pharmacy, Reina Sofia Hospital of Murcia, Murcia, Spain
- Clinical Pharmacokinetics and Applied Pharmacotherapy Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Murcia, Spain
| | - Carles Iniesta-Navalón
- Department of Hospital Pharmacy, Reina Sofia Hospital of Murcia, Murcia, Spain.
- Clinical Pharmacokinetics and Applied Pharmacotherapy Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Murcia, Spain.
- Department of Pharmacology School of Medicine, University of Murcia, Campus de Ciencias de la Salud, Murcia, 30120, Spain.
| | - Elena Urbieta-Sanz
- Department of Hospital Pharmacy, Reina Sofia Hospital of Murcia, Murcia, Spain
- Clinical Pharmacokinetics and Applied Pharmacotherapy Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Murcia, Spain
- Department of Pharmacology School of Medicine, University of Murcia, Campus de Ciencias de la Salud, Murcia, 30120, Spain
| | - Juan José Gascón-Cánovas
- Department of Public Health, University of Murcia, Campus de Ciencias de la Salud, Murcia, 30120, Spain
| |
Collapse
|
3
|
Gehin JE, Klaasen RA, Klami Kristianslund E, Jyssum I, Sexton J, Warren DJ, Aletaha D, Haavardsholm EA, Syversen SW, Goll GL, Bolstad N. Therapeutic serum level for adalimumab in rheumatoid arthritis: explorative analyses of data from a randomised phase III trial. RMD Open 2024; 10:e004888. [PMID: 39537558 PMCID: PMC11575345 DOI: 10.1136/rmdopen-2024-004888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES The objectives of this study are to identify a therapeutic serum level for adalimumab associated with remission and low disease activity in patients with rheumatoid arthritis. METHODS Associations between serum adalimumab trough levels and disease activity were examined using longitudinal data from a 48-week randomised phase III trial including patients with tumour necrosis factor inhibitor-naïve rheumatoid arthritis with active disease starting adalimumab treatment. Disease activity was classified according to 28-joint Disease Activity Score (DAS28)-erythrocyte sedimentation rate and C reactive protein (CRP) levels. RESULTS Adalimumab trough levels were recorded longitudinally for 336, 330 and 302 patients at weeks 12, 24 and 48, respectively. All patients received concomitant methotrexate. Median adalimumab trough levels were 6.4 mg/L (IQR 3.4-9.5) at week 12, 7.5 mg/L (IQR 3.5-10.9) at week 24 and 7.6 mg/L (IQR 3.6-12.0) at week 48. In serial serum samples from weeks 12, 24 and 48, trough levels ≥3.9 mg/L were associated with DAS28 remission (OR 3.88 (95% CI 1.80, 8.38), p<0.001) and lower CRP levels (p<0.001). Week 12 trough levels ≥3.5 mg/L were associated with DAS28 low disease activity at week 24 (OR 2.62 (1.50, 4.56), p<0.001) and remission at week 48 (OR 1.99 (1.02, 3.88), p=0.04), as well as lower CRP levels at both time points (p<0.001). CONCLUSION Adalimumab trough levels above 4.0 mg/L were associated with remission/low disease activity throughout the first year of adalimumab therapy and can be considered a lower target level for therapeutic drug monitoring of adalimumab therapy.
Collapse
Affiliation(s)
- Johanna Elin Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Rolf Anton Klaasen
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Eirik Klami Kristianslund
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Ingrid Jyssum
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Joseph Sexton
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - David John Warren
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Daniel Aletaha
- Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Espen Andre Haavardsholm
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Silje Watterdal Syversen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Guro Løvik Goll
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nils Bolstad
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
4
|
Hooijberg F, van den Berg SPH, Layegh Z, Leeuw M, Elkayam O, de Vries A, Nurmohamed M, Rispens T, Dorlo TPC, Wolbink G. Precision Dosing of Intravenous Tocilizumab: Development of Pharmacokinetic Model-Derived Tapering Strategies for Patients With Rheumatoid Arthritis. Ther Drug Monit 2024:00007691-990000000-00276. [PMID: 39509293 DOI: 10.1097/ftd.0000000000001258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/05/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Tocilizumab targets the interleukin-6 receptor, and dosing is complex owing to its nonlinear clearance related to target binding. Therefore, tapering tocilizumab requires a different approach than that of tumor necrosis factor inhibitors (TNFi). This study aimed to identify these differences and enable personalized treatment of rheumatoid arthritis (RA) beyond TNFi therapy. METHODS A population pharmacokinetic model of intravenous tocilizumab was developed using data from a randomized controlled trial of dose tapering in patients with RA. Subsequent population-level Monte Carlo and individual Bayesian simulations were performed to create tapering strategies involving dose reduction and interval extension. The target trough concentration of tocilizumab was 5 mg/L. Finally, the drug savings were compared between the 2 methods. RESULTS The pharmacokinetic of tocilizumab was described with a 2-compartment model with parallel linear (CL 0.20 L/d) and nonlinear (VM 5.2 mg/d, KM 0.19 mg/L) elimination. The linear clearance rate and central volume of distribution increased with lean body mass, and men exhibited higher clearance rates than women. The simulated concentration-time profiles demonstrated that, owing to nonlinear clearance, drug concentrations decreased more than dose-proportionally with lower doses. Tapering based on an individual Bayesian approach emerged as the most promising strategy, yielding a 39% reduction in drug use across virtual populations. CONCLUSIONS Tapering strategies were developed for intravenous tocilizumab, offering potential application in patients with RA who have reached low disease activity or remission, pending clinical validation. The developed strategies demonstrate that the tapering of tocilizumab should be approached more carefully and in smaller steps than that of TNFi.
Collapse
Affiliation(s)
- Femke Hooijberg
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam, the Netherlands
- Department of Rheumatology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Stefan P H van den Berg
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - Zohra Layegh
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Maureen Leeuw
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam, the Netherlands
- Department of Rheumatology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ori Elkayam
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel
| | - Annick de Vries
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
- R&D, Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Mike Nurmohamed
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam, the Netherlands
- Department of Rheumatology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | | | - Gertjan Wolbink
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam, the Netherlands
- Department of Rheumatology, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Uhrenholt L, Sørensen MER, Lauridsen KB, Duch K, Dreyer L, Christensen R, Hauge EM, Loft AG, Rasch MNB, Horn HC, Taylor PC, Nielsen KR, Kristensen S. Exploring TNFi drug-levels and anti-drug antibodies during tapering among patients with inflammatory arthritis: secondary analyses from the randomised BIODOPT trial. Rheumatol Int 2024; 44:1897-1908. [PMID: 39043980 PMCID: PMC11392959 DOI: 10.1007/s00296-024-05665-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
To evaluate tumour necrosis factor inhibitor (TNFi) drug-levels and presence of anti-drug antibodies (ADAb) in patients with inflammatory arthritis who taper TNFi compared to TNFi continuation. Patients with rheumatoid arthritis, psoriatic arthritis, or axial spondyloarthritis on stable TNFi dose and in low disease activity ≥ 12 months were randomised (2:1) to disease activity-guided tapering or control. Blood samples at baseline, 12- and 18-months were evaluated for TNFi drug-levels and ADAb. In total, 129 patients were randomised to tapering (n = 88) or control (n = 41). Between baseline and month 18, a significant shift in TNFi drug-levels were observed in the tapering group resulting in fewer patients with high drug-levels (change: - 14% [95% CI - 27 to - 1%]) and more with low drug-levels (change: 18% [95% CI 5-31%]). Disease activity was equivalent between groups at 18 months, mean difference: RA - 0.06 (95% CI - 0.44 to 0.33), PsA 0.03 (95% CI - 0.36 to 0.42), and axSpA 0.16 (- 0.17 to 0.49), equivalence margins ± 0.5 disease activity points. ADAb were detected in eight patients, all from the tapering group. TNFi drug-level category or ADAb were not predictive for achieving successful tapering at 18 months. TNFi drug-levels decreased during tapering which indicate adherence to the tapering algorithm. Despite the difference in TNFi drug-levels at 18 months, disease activity remained equivalent, and only few tapering patients had detectable ADAb. These data do not support using TNFi drug-level and/or ADAb to guide the tapering decision but future research with larger trials is needed.Trial registration: EudraCT: 2017-001970-41, December 21, 2017.
Collapse
Affiliation(s)
- Line Uhrenholt
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark.
| | - Mads E R Sørensen
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Karen B Lauridsen
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Kirsten Duch
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark
- Research Data and Biostatistics, Aalborg University Hospital, Aalborg, Denmark
| | - Lene Dreyer
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Robin Christensen
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Ellen-Margrethe Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne Gitte Loft
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mads N B Rasch
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Kaspar R Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Salome Kristensen
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
6
|
Li Y, Xie C, Ding X, Wu Z, Zhang J, Zhu J, Miao L. What are the benefits of therapeutic drug monitoring in the optimization of adalimumab therapy? a systematic review and meta-analysis up to 2022. Front Pharmacol 2024; 15:1376708. [PMID: 39040471 PMCID: PMC11260779 DOI: 10.3389/fphar.2024.1376708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Aims Persistent uncertainties exist surrounding the therapeutic drug monitoring (TDM) of adalimumab in clinical settings. To address these issues, we conducted a systematic review to assess the current evidence regarding the benefits of TDM for adalimumab. Methods PubMed, EMBASE, and Cochrane Databases were searched from inception to October 2022. The trials regarding to the list three key questions were considered: 1) Could routine proactive TDM assist in improving outcomes in patients receiving adalimumab? 2) Could reactive TDM assist in guiding subsequent treatment strategies for patients with treatment failure to adalimumab? 3) Could TDM assist in informing dose reduction or discontinuation in patients with low disease activity or in remission treated with adalimumab? Two reviewers independently selected the studies and extracted the data. Meta-analysis was performed to calculate the relative risk (RR) and 95% confidence interval (CI). Results A total of 9 studies was included in this review. For proactive TDM, meta-analysis indicated that proactive TDM (n = 163/257, 63.42%) showed no significant superiority over reactive TDM and/or conventional management (n = 336/606, 55.44%) in achieving and/or maintaining clinical remission by random effects model (RR: 1.24, 95% CI 0.98-1.58, I 2 = 73%). There were three studies that supporting the reactive TDM, low drug levels in the absence of anti-drug antibodies (ADA) strongly indicate the need for dose intensification, and infliximab is a feasible choice for patients with low drug levels and ADA positivity. While swapping to another class should be considered in patients with adequate drug levels. In addition, TDM can help clinicians optimize dosing schedules and prevent overtreatment in patients who have achieved low disease activity and sufficient drug concentrations, with no predictive value for successful adalimumab discontinuation. Conclusion Current evidence suggests that proactive TDM is numerically but not statistically significant superiority over reactive TDM and/or conventional management. Reactive TDM can aid in understanding treatment failure and developing subsequent therapy. For patients reaching low disease activity and remission, TDM can help successful dose reduction, while it cannot inform the successful drug discontinuation. However, existing trials are limited, and more well-designed trials are necessary to clarify the role of TDM in adalimumab treatment.
Collapse
Affiliation(s)
- Yun Li
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Xie
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoliang Ding
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziyang Wu
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingjing Zhang
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianguo Zhu
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Chen L, Kang D, Fang L, Sun M, Li M, Zhou G, Xu C, Pang Z, Ye Y, Feng B, Wu H, Lin J, Ding B, Liu C, Shi Y, Liu Z. Development and validation of a novel therapeutic drug monitoring-based nomogram for prediction of primary endoscopic response to anti-TNF therapy in active Crohn's disease. Therap Adv Gastroenterol 2024; 17:17562848241256237. [PMID: 38827646 PMCID: PMC11143805 DOI: 10.1177/17562848241256237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/04/2024] [Indexed: 06/04/2024] Open
Abstract
Background Anti-tumor necrosis factor (TNF) monoclonal antibodies, especially infliximab (IFX) and adalimumab (ADA), are considered the first-line treatment for active Crohn's disease (CD). However, the predictive role of therapeutic drug monitoring (TDM) of serum anti-TNF in monitoring the treatment of inflammatory bowel disease (IBD) remains controversial. Objectives To explore the correlation between serum anti-TNF levels and early endoscopic response in active CD using a TDM-based nomogram. Design Cross-sectional study. Methods The simplified endoscopic activity score for CD (SES-CD), Crohn's disease activity index (CDAI), laboratory parameters, and the serum trough levels of IFX and ADA were assessed. Results The trough levels of IFX or ADA were significantly higher in patients with endoscopic response compared to non-responders in the development cohort (p < 0.001). The IFX and ADA levels showed a weak but significantly negative correlation with SES-CD (p < 0.001), CDAI (p < 0.001), and C-reactive protein (CRP) (p < 0.001) at week 14 post-IFX therapy in the development cohort. Furthermore, the receiver operating characteristic curve revealed that an optimal level of IFX (4.80 μg/mL) and ADA (8.80 μg/mL) exhibited the best performance in predicting endoscopic response. Concomitantly, we developed a novel nomogram prediction model based on the results of multivariate logistic regression analysis, which consisted of CRP, albumin (Alb), and anti-TNF trough levels at week 14. The nomogram showed significant discrimination and calibration for both IFX and ADA in the development cohort and performed well in the external validation cohort. Conclusion This study demonstrates a robust association between serum concentrations of IFX, ADA, Alb, and CRP and primary endoscopic response in active CD patients. Importantly, the TDM- and laboratory marker-based nomogram may be used to evaluate the primary endoscopic response to anti-TNF therapy, especially for optimizing treatment strategies and switching therapy in CD patients.
Collapse
Affiliation(s)
- Liang Chen
- Department of Gastroenterology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Dengfeng Kang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Leilei Fang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Mingming Sun
- Department of Gastroenterology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Mingsong Li
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical College, Jining, China
| | - Chunjin Xu
- Department of Gastroenterology, First People’s Hospital of Shangqiu City Affiliated to Xinxiang Medical University, Shangqiu, China
| | - Zhi Pang
- Department of Gastroenterology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Yulan Ye
- Department of Gastroenterology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Baisui Feng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huili Wu
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jian Lin
- Department of Gastroenterology, Affiliated Hospital of Putian University, Putian, China
| | - Baijing Ding
- Department of Gastroenterology, Wuhu First People’s Hospital, Wuhu, Anhui, China
| | - Changqin Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai 200072, China
| | - Yanhong Shi
- Department of Gastroenterology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai 200072, China
| | - Zhanju Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai 200072, China
| |
Collapse
|
8
|
Stamp LK, Keating P, Frampton C, Barclay ML, Fanning N, Millier M, Hessian PA, O'Donnell JL. Relationship Between Adalimumab Concentrations, Antidrug Antibodies, and Disease Activity in Rheumatoid Arthritis: A Cross-Sectional Observational Study. J Rheumatol 2024; 51:242-249. [PMID: 38224989 DOI: 10.3899/jrheum.2023-0706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/17/2024]
Abstract
OBJECTIVES To determine the influence of patient characteristics and disease activity on adalimumab (ADA) concentrations; to assess the relationships between ADA concentrations, the presence of antidrug antibodies (ADAb), and disease activity in rheumatoid arthritis (RA); and to determine the association between cytokine concentrations and ADA concentrations. METHODS A cross-sectional study of people with RA receiving ADA for at least 4 weeks was undertaken. Disease activity was assessed by the Disease Activity Score in 28 joints (DAS28), with responders defined as DAS28 ≤ 3.2. Serum and plasma were obtained for ADA concentrations and ADAb, and a panel of cytokines were obtained for a subgroup. ADA concentrations were compared between demographic and clinical subgroups using ANOVA. The independent associations between clinical and demographic features were analyzed using a general linear model. Variables significantly associated with ADA concentrations from the univariate analyses were entered into multivariate analyses. RESULTS Of the 156 participants, 69.2% were female and the mean age was 57.4 (SD 12.7) years. Multivariate analysis revealed that higher C-reactive protein (P < 0.001) and higher weight (P < 0.004) were independently associated with lower ADA concentrations. ADA concentrations were higher in those with DAS28 ≤ 3.2 compared to those with DAS28 > 3.2 (median 10.8 [IQR 6.4-20.8] mg/L vs 7.1 [IQR 1.5-12.6] mg/L, P < 0.001). There was a significant negative correlation between interleukin 6 (IL-6) and ADA concentrations (r = -0.04, P < 0.01). CONCLUSION ADA concentration correlates negatively with markers of inflammatory disease activity in RA, including IL-6. ADA concentration in the range 5 to 7 mg/L over the dose interval are associated with better disease control.
Collapse
Affiliation(s)
- Lisa K Stamp
- L.K. Stamp, MBChB, PhD, C. Frampton, PhD, M.L. Barclay, MBChB, N. Fanning, PhD, Department of Medicine, University of Otago Christchurch, Christchurch;
| | - Paula Keating
- P. Keating, PhD, J.L. O'Donnell, FRACP, Canterbury Health Laboratories, Te Whatu Ora, Waitaha
| | - Christopher Frampton
- L.K. Stamp, MBChB, PhD, C. Frampton, PhD, M.L. Barclay, MBChB, N. Fanning, PhD, Department of Medicine, University of Otago Christchurch, Christchurch
| | - Murray L Barclay
- L.K. Stamp, MBChB, PhD, C. Frampton, PhD, M.L. Barclay, MBChB, N. Fanning, PhD, Department of Medicine, University of Otago Christchurch, Christchurch
| | - Niamh Fanning
- L.K. Stamp, MBChB, PhD, C. Frampton, PhD, M.L. Barclay, MBChB, N. Fanning, PhD, Department of Medicine, University of Otago Christchurch, Christchurch
| | - Melanie Millier
- M. Millier, BSc, P.A. Hessian, PhD, Department of Medicine, University of Otago Dunedin, Dunedin, New Zealand
| | - Paul A Hessian
- M. Millier, BSc, P.A. Hessian, PhD, Department of Medicine, University of Otago Dunedin, Dunedin, New Zealand
| | - John L O'Donnell
- P. Keating, PhD, J.L. O'Donnell, FRACP, Canterbury Health Laboratories, Te Whatu Ora, Waitaha
| |
Collapse
|
9
|
Welzel T, Golhen K, Atkinson A, Gotta V, Ternant D, Kuemmerle-Deschner JB, Michler C, Koch G, van den Anker JN, Pfister M, Woerner A. Prospective study to characterize adalimumab exposure in pediatric patients with rheumatic diseases. Pediatr Rheumatol Online J 2024; 22:5. [PMID: 38167019 PMCID: PMC10763375 DOI: 10.1186/s12969-023-00930-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND In pediatric rheumatic diseases (PRD), adalimumab is dosed using fixed weight-based bands irrespective of methotrexate co-treatment, disease activity (DA) or other factors that might influence adalimumab pharmacokinetics (PK). In rheumatoid arthritis (RA) adalimumab exposure between 2-8 mg/L is associated with clinical response. PRD data on adalimumab is scarce. Therefore, this study aimed to analyze adalimumab PK and its variability in PRD treated with/without methotrexate. METHODS A two-center prospective study in PRD patients aged 2-18 years treated with adalimumab and methotrexate (GA-M) or adalimumab alone (GA) for ≥ 12 weeks was performed. Adalimumab concentrations were collected 1-9 (maximum concentration; Cmax), and 10-14 days (minimum concentration; Cmin) during ≥ 12 weeks following adalimumab start. Concentrations were analyzed with enzyme-linked immunosorbent assay (lower limit of quantification: 0.5 mg/L). Log-normalized Cmin were compared between GA-M and GA using a standard t-test. RESULTS Twenty-eight patients (14 per group), diagnosed with juvenile idiopathic arthritis (71.4%), non-infectious uveitis (25%) or chronic recurrent multifocal osteomyelitis (3.6%) completed the study. GA-M included more females (71.4%; GA 35.7%, p = 0.13). At first study visit, children in GA-M had a slightly longer exposure to adalimumab (17.8 months [IQR 9.6, 21.6]) compared to GA (15.8 months [IQR 8.5, 30.8], p = 0.8). Adalimumab dosing was similar between both groups (median dose 40 mg every 14 days) and observed DA was low. Children in GA-M had a 27% higher median overall exposure compared to GA, although median Cmin adalimumab values were statistically not different (p = 0.3). Cmin values ≥ 8 mg/L (upper limit RA) were more frequently observed in GA-M versus GA (79% versus 64%). Overall, a wide range of Cmin values was observed in PRD (0.5 to 26 mg/L). CONCLUSION This study revealed a high heterogeneity in adalimumab exposure in PRD. Adalimumab exposure tended to be higher with methotrexate co-treatment compared to adalimumab monotherapy although differences were not statistically significant. Most children showed adalimumab exposure exceeding those reported for RA with clinical response, particularly with methotrexate co-treatment. This highlights the need of further investigations to establish model-based personalized treatment strategies in PRD to avoid under- and overexposure. TRIAL REGISTRATION NCT04042792 , registered 02.08.2019.
Collapse
Affiliation(s)
- Tatjana Welzel
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland.
- Division of Pediatric Rheumatology, Department of Pediatrics and Autoinflammation Reference Centre Tuebingen (arcT), University Hospital Tuebingen, Tuebingen, Germany.
- Pediatric Rheumatology, University Children's Hospital Basel, University of Basel, Basel, Switzerland.
- Pediatric Research Center, University Children's Hospital Basel, University of Basel, Basel, Switzerland.
| | - Klervi Golhen
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrew Atkinson
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Pediatric Research Center, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Division of Infectious Diseases, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Verena Gotta
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - David Ternant
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Université de Tours, service de pharmacologie médicale, Tours France, Université de Tours, EA 4245 T2I, Tours, France
| | - Jasmin B Kuemmerle-Deschner
- Division of Pediatric Rheumatology, Department of Pediatrics and Autoinflammation Reference Centre Tuebingen (arcT), University Hospital Tuebingen, Tuebingen, Germany
| | - Christine Michler
- Division of Pediatric Rheumatology, Department of Pediatrics and Autoinflammation Reference Centre Tuebingen (arcT), University Hospital Tuebingen, Tuebingen, Germany
| | - Gilbert Koch
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Johannes N van den Anker
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Andreas Woerner
- Pediatric Rheumatology, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Dong W, Hu X, Wu C, Wang G, Fang Y, Shi L, Nie X. Efficacy, safety, and cost-effectiveness of therapeutic drug monitoring (TDM) for TNF inhibitor therapy in rheumatic disease: A systematic review and meta-analysis. Semin Arthritis Rheum 2023; 63:152302. [PMID: 37951128 DOI: 10.1016/j.semarthrit.2023.152302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/13/2023]
Abstract
OBJECTIVE The benefits of TDM-guided TNFi therapy in patients with rheumatic disease was still controversial. This systematic review and meta-analysis was conducted to explore if the TDM-guided TNFi therapy is superior to empirical-guided therapy. METHODS We systematically searched PubMed, Web of Science, Cochrane Library, and EMBASE databases for articles published between database inception and October 05, 2023. Studies reporting endpoints in TDM-guided TNFi therapy and empirical therapy were included. Results would be presented in risk ratio (RR) and mean difference, with 95 % confidence interval (CI) reported. This study is registered with PROSPERO (CRD42022353956). RESULTS A total of 14 studies (eight RCTs and six cohort studies) involving 2427 patients were included in this meta-analysis. In the scenario of response prediction, compared with empirical-guided therapy, TDM-guided TNFi therapy had association with higher treat-to-target rates (RR 1.30, 95 % CI 1.02-1.65, P=0.03, I2=79 %), more specifically, higher low disease activity rates (RR 2.11, 95 % CI 1.22-3.66, P=0.007, I2=61 %), but no difference in clinical remission rates (RR 0.98,95 % CI 0.87-1.11, P=0.75, I2=0 %). In the scenario of dose reduction prediction, lower relapse rates (RR 0.73, 95 % CI 0.65-0.82, P <0.00001, I2=0 %) were observed compared with empirical-guided dose reduction strategy, but no difference (RR 1.24, 95 % CI 0.85-1.80, P=0.27, I2=57 %) between TDM-guided dose reduction and standard-dosing therapy. No significant difference was observed in change of disease activity score, mean disease activity score, radiographic progression, and safety. And TDM-guided therapy was associated with reduced cost per patient per year calculated as the total accumulated sum of therapy cost. CONCLUSION TDM-guided TNFi therapy was associated with increased rates of low disease activity and decreased risks of relapse, and may save cost compared with empirical-guided therapy in patients with rheumatic disease. But this does not mean that the use of TDM-guided TNFi therapy can be advocated, because there is no difference in clinical remission rates and many other outcomes. More researches, especially randomized clinical trials are needed to verify this conclusion in the future.
Collapse
Affiliation(s)
- Wenliang Dong
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Clinical Trial Institution, Peking University People's Hospital, Beijing 101109, China
| | - Xiaowen Hu
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Caiying Wu
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Gengchen Wang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yi Fang
- Clinical Trial Institution, Peking University People's Hospital, Beijing 101109, China.
| | - Luwen Shi
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; International Research Center for Medicinal Administration, Peking University, Beijing 100191, China.
| | - Xiaoyan Nie
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; International Research Center for Medicinal Administration, Peking University, Beijing 100191, China.
| |
Collapse
|
11
|
Raharja A, Arkir Z, Rinaldi G, Tsakok T, Dasandi T, Guard S, McGuire A, Pink AE, Woolf R, Barker JN, Smith CH, Mahil SK. Real-World Implementation and Outcomes of Adalimumab Therapeutic Drug Monitoring in Psoriasis: A National Specialized Center Experience. J Invest Dermatol 2023; 143:1708-1716.e4. [PMID: 36889663 DOI: 10.1016/j.jid.2023.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 03/08/2023]
Abstract
Serum adalimumab concentration is a biomarker of treatment response but therapeutic drug monitoring (TDM) is yet to be implemented in routine psoriasis care. We incorporated adalimumab TDM in a national specialized psoriasis service and evaluated it using the RE-AIM (Reach, Effectiveness, Adoption, Implementation, and Maintenance) implementation science framework. We undertook pre-implementation planning (validating local assays) and implementation interventions targeted to patients (pragmatic sampling at routine reviews), clinicians (introduction of a TDM protocol), and healthcare systems (adalimumab TDM as a key performance indicator). Over 5 months, 170 of 229 (74%) individuals treated with adalimumab received TDM. Clinical improvement after TDM-guided dose escalation occurred in 13 of 15 (87%) nonresponders with serum drug concentrations <8.3 μg/ml (median PASI reduction of 3.2 [interquartile range = 2.2-8.2] after 23.4 weeks) and in all nonresponders who had TDM-guided switch in biologic due to supratherapeutic drug concentrations (>8.3 μg/ml; n = 2) or positive antidrug antibody (n = 2) (PASI reduction of 7.8 [interquartile range = 7.5-12.9] after 20.0 weeks). Proactive TDM led to dose reduction in five individuals with clear skin and subtherapeutic or supratherapeutic drug concentrations; four (80%) sustained clear skin after 50 weeks (range = 42-52). Adalimumab TDM based on pragmatic serum sampling is clinically viable and may lead to patient benefit. Context-specific implementation interventions and systematic implementation assessment may bridge the biomarker research-to-practice gap.
Collapse
Affiliation(s)
- Antony Raharja
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Zehra Arkir
- Reference Chemistry, Synnovis, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Giulia Rinaldi
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Teresa Tsakok
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Tejus Dasandi
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Sarah Guard
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Arlene McGuire
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Andrew E Pink
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Richard Woolf
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Jonathan N Barker
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Catherine H Smith
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Satveer K Mahil
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom.
| |
Collapse
|
12
|
Krieckaert CL, van Tubergen A, Gehin JE, Hernández-Breijo B, Le Mélédo G, Balsa A, Böhm P, Cucnik S, Elkayam O, Goll GL, Hooijberg F, Jani M, Kiely PD, McCarthy N, Mulleman D, Navarro-Compán V, Payne K, Perry ME, Plasencia-Rodriguez C, Stones SR, Syversen SW, de Vries A, Ward KM, Wolbink G, Isaacs JD. EULAR points to consider for therapeutic drug monitoring of biopharmaceuticals in inflammatory rheumatic and musculoskeletal diseases. Ann Rheum Dis 2023; 82:65-73. [PMID: 35551063 DOI: 10.1136/annrheumdis-2022-222155] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/06/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To develop EULAR points-to-consider for therapeutic drug monitoring (TDM) of biopharmaceuticals in inflammatory rheumatic and musculoskeletal diseases (RMDs). METHODS The points-to-consider were developed in accordance with EULAR standardised operation procedures by a multidisciplinary task force from eight European countries, based on a systematic literature review and expert consensus. Level of evidence and strength of the points-to-consider were determined, and mean levels of agreement among the task force were calculated using a 10-point rating scale. RESULTS Six overarching principles and 13 points-to-consider were formulated. The level of agreement among the task force for the overarching principles and points-to-consider ranged from 8.4 to 9.9.The overarching principles define TDM and its subtypes, and reinforce the underlying pharmacokinetic/pharmacodynamic principles, which are relevant to all biopharmaceutical classes. The points-to-consider highlight the clinical utility of the measurement and interpretation of biopharmaceutical blood concentrations and antidrug antibodies in specific clinical scenarios, including factors that influence these parameters. In general, proactive use of TDM is not recommended but reactive TDM could be considered in certain clinical situations. An important factor limiting wider adoption of TDM is the lack of both high quality trials addressing effectiveness and safety of TDM and robust economic evaluation in patients with RMDs. Future research should focus on providing this evidence, as well as on further understanding of pharmacokinetic and pharmacodynamic characteristics of biopharmaceuticals. CONCLUSION These points-to-consider are evidence-based and consensus-based statements for the use of TDM of biopharmaceuticals in inflammatory RMDs, addressing the clinical utility of TDM.
Collapse
Affiliation(s)
- Charlotte Lm Krieckaert
- Reumatology, Amsterdam Rheumatology and Immunology Center, Reade, Amsterdam, The Netherlands
| | - Astrid van Tubergen
- Medicine, Division of Rheumatology, Maastricht University Medical Centre+, Maastricht, The Netherlands.,CAPHRI, Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Johanna Elin Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | | | - Alejandro Balsa
- Immuno-Rheumatology Research Group, La Paz University Hospital, Madrid, Spain.,Rheumatology, La Paz University Hospital, Madrid, Spain
| | - Peter Böhm
- Forschungspartner, Deutsche Rheuma-Liga Bundesverband, Bonn, Germany
| | - Sasa Cucnik
- Rheumatology, Ljubljanski Univerzitetni klinicni center, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Ori Elkayam
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Guro L Goll
- Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Femke Hooijberg
- Rheumatology, Reade Hoofdlocatie Dr Jan van Breemenstraat, Amsterdam, The Netherlands
| | - Meghna Jani
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,Rheumatology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Patrick Dw Kiely
- Department of Rheumatology, St George's University Hospitals NHS Foundation Trust, London, UK.,Institute of Medical and Biochemical Education, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Neil McCarthy
- Patient Representative, Newcastle upon Tyne Hospitals NHS Foundation Trust, Manchester, UK
| | - Denis Mulleman
- Rheumatology, Regional University Hospital Centre Tours, Tours, France
| | | | - Katherine Payne
- Division of Population Health, Health Services Research and Primary Care, Manchester Academic Health Science Centre, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Martin E Perry
- Centre for Rheumatic Diseases, Royal Alexandra Hospital, Paisley, UK
| | | | - Simon R Stones
- EULAR Patient Research partner, Collaboro Consulting, Manchester, UK
| | | | | | - Katherine M Ward
- Department of Rheumatology, Diakonhjemmet Hospital Department of Rheumatology, Oslo, Norway
| | - Gertjan Wolbink
- Reumatology, Amsterdam Rheumatology and Immunology Center, Reade, Amsterdam, The Netherlands.,Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK .,Musculoskeletal Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| |
Collapse
|
13
|
Gehin JE, Goll GL, Brun MK, Jani M, Bolstad N, Syversen SW. Assessing Immunogenicity of Biologic Drugs in Inflammatory Joint Diseases: Progress Towards Personalized Medicine. BioDrugs 2022; 36:731-748. [PMID: 36315391 PMCID: PMC9649489 DOI: 10.1007/s40259-022-00559-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 11/30/2022]
Abstract
Biologic drugs have greatly improved treatment outcomes of inflammatory joint diseases, but a substantial proportion of patients either do not respond to treatment or lose response over time. Drug immunogenicity, manifested as the formation of anti-drug antibodies (ADAb), constitute a significant clinical problem. Anti-drug antibodies influence the pharmacokinetics of the drug, are associated with reduced clinical efficacy, and an increased risk of adverse events such as infusion reactions. The prevalence of ADAb differs among drugs and diseases, and the detection of ADAb also depends on the assay format. Most data exist for the tumor necrosis factor-alpha inhibitors infliximab and adalimumab, with a frequency of ADAb that ranges from 10 to 60% across studies. Measurement of ADAb and serum drug concentrations, therapeutic drug monitoring, has been suggested as a strategy to optimize therapy with biologic drugs. Although the recent randomized clinical Norwegian Drug Monitoring (NOR-DRUM) trials show promise towards a personalized medicine prescribing approach by therapeutic drug monitoring, several challenges remain. A plethora of assay formats, with widely differing properties, is currently used for measuring ADAb. Comparing results between different assays and laboratories is difficult, which complicates the development of cut-offs necessary for guidelines and the implementation of ADAb measurements in clinical practice. With the possible exception of infliximab, limited data on clinical relevance and cost effectiveness exist to support therapeutic drug monitoring as a routine clinical strategy to monitor biologic drugs in inflammatory joint diseases. The aim of this review is to provide an overview of the characteristics and prevalence of ADAb, predisposing factors to ADAb formation, commonly used assessment methods, clinical consequences of ADAb, and the potential implications of ADAb assessments for everyday treatment of inflammatory joint diseases.
Collapse
Affiliation(s)
- Johanna Elin Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Radiumhospitalet, Nydalen, Box 4953, 0424, Oslo, Norway.
| | - Guro Løvik Goll
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Marthe Kirkesæther Brun
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Meghna Jani
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, UK
- Department of Rheumatology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Nils Bolstad
- Department of Medical Biochemistry, Oslo University Hospital, Radiumhospitalet, Nydalen, Box 4953, 0424, Oslo, Norway
| | - Silje Watterdal Syversen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
14
|
Gao J, Miao J, Sun H, Fu X, Zhang P, Chen Z, Zhu P. TNF-α inhibitor ameliorates immune-related arthritis and pneumonitis in humanized mice. Front Immunol 2022; 13:955812. [PMID: 36016934 PMCID: PMC9396351 DOI: 10.3389/fimmu.2022.955812] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesThis study aimed at establishing a mouse model of immune-related adverse in humanized BALB/c-hPD1/hCTLA4 mice to investigate their potential pathogenesis and explore therapeutic targets for immune-related arthritis and pneumonitis.MethodsHumanized BALB/c-hPD1/hCTLA4 mice were injected with vehicle or collagen-specific antibodies (CA) and immune checkpoint inhibitors (ICI, ipilimumab, anti-human CTLA-4; and nivolumab, anti-human PD-1), and some mice were treated with anti-TNF-α antibody, leading to the control, collagen antibody-induced arthritis (CAIA), CAIA+ICI and treatment groups. The severity of clinical arthritis and pneumonitis in mice was monitored longitudinally and the pathological changes in the joints and lungs were histologically analyzed and the contents of lung hydroxyproline were measured. The frequency of different subsets of T cells was analyzed by flow cytometry and multiplex immunofluorescency.ResultsCompared with the control, the ICI group of mice developed the delayed onset of moderate degrees of arthritis while the CAIA+ICI group of mice exhibited the early onset of severe arthritis. Treatment with ICI caused severe pneumonitis, especially in the mice with CA. Flow cytometry analysis indicated a significantly higher frequency of splenic TNF-α+CD4+ and TNF-α+CD8+ T cells, but not other subsets of T cells tested, in the CAIA+ICI group of mice, relative to that in other groups of mice. Treatment with anti-TNF-α significantly mitigated the severity of arthritis and pneumonitis as well as deposition of collagen in lung of mice. The treatment also decreased the frequency of TNF-α+CD4+ and TNF-α+CD8+ T cells as well as effector memory T cells in the periphery lymph orangs and lungs of mice.ConclusionsWe successfully established a humanized mouse model of ICI-related severe arthritis and pneumonitis with a higher frequency of TNF-α+ T cells, which were significantly mitigated by anti-TNF-α treatment. Conceptually, ICI treatment can induce multiple autoimmune-like diseases in autoimmune-prone individuals and TNF-α+ T cells may be therapeutic targets for intervention of immune-related arthritis and pneumonitis.
Collapse
Affiliation(s)
- Jian Gao
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Jinlin Miao
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Ping Zhu, ; Zhinan Chen, ; Jinlin Miao,
| | - Haoyang Sun
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xianghui Fu
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Peiyan Zhang
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhinan Chen
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
- *Correspondence: Ping Zhu, ; Zhinan Chen, ; Jinlin Miao,
| | - Ping Zhu
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Ping Zhu, ; Zhinan Chen, ; Jinlin Miao,
| |
Collapse
|
15
|
Krieckaert C, Hernández-Breijo B, Gehin JE, le Mélédo G, Balsa A, Jani M, Mulleman D, Navarro-Compan V, Wolbink G, Isaac J, van Tubergen A. Therapeutic drug monitoring of biopharmaceuticals in inflammatory rheumatic and musculoskeletal disease: a systematic literature review informing EULAR points to consider. RMD Open 2022; 8:e002216. [PMID: 35980738 PMCID: PMC9171282 DOI: 10.1136/rmdopen-2022-002216] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/18/2022] [Indexed: 01/08/2023] Open
Abstract
The objectives of this review were to collect and summarise evidence on therapeutic drug monitoring (TDM) of biopharmaceuticals in inflammatory rheumatic and musculoskeletal diseases and to inform the EULAR Task Force for the formulation of evidence-based points to consider. A systematic literature review (SLR) was performed, covering technical aspects and (clinical) utility of TDM, to answer 13 research questions. MEDLINE, Embase and Cochrane were searched until July 2020. American College of Rheumatology and EULAR abstracts were also considered for inclusion. Data were extracted in evidence tables and risk of bias assessment was performed. For the search on technical aspects, 678 records were identified, of which 22 papers were selected. For the clinical utility search, 3846 records were identified, of which 108 papers were included. Patient-related factors associated with biopharmaceutical blood concentrations included body weight, methotrexate comedication and disease activity. The identification of a target range was hampered by study variability, mainly disease activity measures and study type. Evidence was inconsistent for multiple clinical situations in which TDM is currently applied. However, for some particular scenarios, including prediction of future treatment response, non-response to treatment, tapering and hypersensitivity reactions, robust evidence was found. There is currently no evidence for routine use of proactive TDM, in part because published cost-effectiveness analyses do not incorporate the current landscape of biopharmaceutical costs and usage. This SLR yields evidence in favour of TDM of biopharmaceuticals in some clinical scenarios, but evidence is insufficient to support implementation of routine use of TDM.
Collapse
Affiliation(s)
- Charlotte Krieckaert
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Reade, Amsterdam, The Netherlands
| | | | - Johanna Elin Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | | | | | - Meghna Jani
- Centre for Epidemiology versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
- department of Rheumatology, Salford Royal Hospitals NHS Trust, Salford, UK
| | | | | | - Gertjan Wolbink
- Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - John Isaac
- Translational and Clinical Research Institute, Newcastle University and Musculoskeletal Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Astrid van Tubergen
- department of Medicine, Division of Rheumatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
16
|
Papamichael K, Afif W, Drobne D, Dubinsky MC, Ferrante M, Irving PM, Kamperidis N, Kobayashi T, Kotze PG, Lambert J, Noor NM, Roblin X, Roda G, Vande Casteele N, Yarur AJ, Arebi N, Danese S, Paul S, Sandborn WJ, Vermeire S, Cheifetz AS, Peyrin-Biroulet L. Therapeutic drug monitoring of biologics in inflammatory bowel disease: unmet needs and future perspectives. Lancet Gastroenterol Hepatol 2022; 7:171-185. [PMID: 35026171 PMCID: PMC10187071 DOI: 10.1016/s2468-1253(21)00223-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 01/05/2023]
Abstract
Therapeutic drug monitoring (TDM) has emerged as a useful tool for optimising the use of biologics, and in particular anti-tumour necrosis factor (anti-TNF) therapy, in inflammatory bowel disease (IBD). However, challenges remain and are hindering the widespread implementation of TDM in clinical practice. These barriers include identification of the optimal drug concentration to target, the lag time between sampling and results, and the proper interpretation of anti-drug antibody titres among different assays. Solutions to overcome these barriers include the harmonisation of TDM assays and the use of point-of-care testing. Other unmet needs include well designed prospective studies and randomised controlled trials focusing on proactive TDM, particularly during induction therapy. Future studies should also investigate the utility of TDM for biologics other than anti-TNF therapies in both IBD and other immune-mediated inflammatory diseases such as rheumatoid arthritis and psoriasis, and the use of pharmacokinetic modelling dashboards and pharmacogenetics towards individual personalised medicine.
Collapse
Affiliation(s)
- Konstantinos Papamichael
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Waqqas Afif
- Department of Medicine, Division of Gastroenterology, McGill University Health Center, Montreal, QC, Canada
| | - David Drobne
- Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana, Slovenia; Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Marla C Dubinsky
- Department of Pediatrics, Susan and Leonard Feinstein IBD Clinical Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marc Ferrante
- KU Leuven, Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Peter M Irving
- Gastroenterology, Guy's and St Thomas' Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Paulo G Kotze
- Catholic University of Paraná (PUCPR), Curitiba, Brazil
| | - Jo Lambert
- Department of Head and Skin, Ghent University Hospital, Ghent, Belgium
| | - Nurulamin M Noor
- Department of Gastroenterology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Xavier Roblin
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Giulia Roda
- IBD Center, Humanitas Clinical and Research Center-IRCCS, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | | | - Naila Arebi
- Department of IBD, St Mark's Hospital, Imperial College London, London, UK
| | - Silvio Danese
- IBD Center, Humanitas Clinical and Research Center-IRCCS, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Stephane Paul
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - William J Sandborn
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Séverine Vermeire
- KU Leuven, Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Adam S Cheifetz
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, Nancy, France; INSERM U1256 NGERE, Lorraine University, Nancy, France
| |
Collapse
|
17
|
Uhrenholt L, Christensen R, Dinesen WKH, Liboriussen CH, Andersen SS, Dreyer L, Schlemmer A, Hauge EM, Skrubbeltrang C, Taylor PC, Kristensen S. Risk of flare after tapering or withdrawal of b-/tsDMARDs in patients with RA or axSpA: A systematic review and meta-analysis. Rheumatology (Oxford) 2021; 61:3107-3122. [PMID: 34864896 DOI: 10.1093/rheumatology/keab902] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To evaluate flare risk when tapering or withdrawing biological or targeted synthetic disease-modifying antirheumatic drugs (b-/tsDMARDs) compared to continuation in patients with inflammatory arthritis (IA) in sustained remission or low disease activity. METHODS Articles were identified in Cochrane Library, PubMed, EMBASE and Web of Science. Eligible trials were randomised, controlled trials comparing tapering and/or withdrawal of b- and/or tsDMARDs with standard dose in IA. Random-effects meta-analysis was performed with risk ratio (RR), or Peto's Odds Ratio (POR) for sparse events, and 95% confidence intervals (95%CI). RESULTS The meta-analysis comprised 22 trials: 11 assessed tapering and 7 addressed withdrawal (4 assessed both). Only trials with a rheumatoid arthritis (RA) or axial spondyloarthritis (axSpA) population were identified. An increased flare risk was demonstrated when b-/tsDMARD tapering was compared to continuation, RR = 1.45 (95%CI: 1.19 to 1.77, I2 = 42.5%), and potentially increased for persistent flare, POR = 1.56 (95%CI: 0.97 to 2.52, I2 = 0%). Comparing tumour necrosis factor inhibitor (TNFi) withdrawal to continuation, a highly increased flare risk (RR = 2.28, 95%CI: 1.78 to 2.93, I2 = 78%) and increased odds of persistent flare (POR = 3.41, 95%CI: 1.91 to 6.09, I2 = 49%) was observed. No clear difference in flare risk between RA or axSpA was observed. CONCLUSION A high risk for flare and persistent flare was demonstrated for TNFi withdrawal whereas an increased risk for flare but not for persistent flare was observed for b-/tsDMARD tapering. Thus, tapering seems to be the more favourable approach. REGISTRATION PROSPERO (CRD42019136905).
Collapse
Affiliation(s)
- Line Uhrenholt
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Section for Biostatistics and Evidence-Based Research, Bispebjerg and Frederiksberg Hospital, The Parker Institute, Copenhagen, Denmark
| | - Robin Christensen
- Section for Biostatistics and Evidence-Based Research, Bispebjerg and Frederiksberg Hospital, The Parker Institute, Copenhagen, Denmark.,Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | | | | | - Stine S Andersen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Lene Dreyer
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Annette Schlemmer
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Rheumatology, Randers Regional Hospital, Randers, Denmark
| | - Ellen-Margrethe Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Conni Skrubbeltrang
- Department of Medical Library, Aalborg University Hospital, Aalborg, Denmark
| | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Salome Kristensen
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
18
|
Berkhout LC, l'Ami MJ, Wolbink GJ, Rispens T. Comment on 'Sustained discontinuation of infliximab with a raising-dose strategy after obtaining remission in patients with rheumatoid arthritis: the RRRR study, a randomised controlled trial' by Tanaka et al. Ann Rheum Dis 2021; 80:e172. [PMID: 31744825 DOI: 10.1136/annrheumdis-2019-216557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 11/04/2022]
Affiliation(s)
- Lea C Berkhout
- Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - Merel J l'Ami
- Reade, Amsterdam Rheumatology and immunology Center, Amsterdam, The Netherlands
| | - Gerrit Jan Wolbink
- Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Reade, Amsterdam Rheumatology and immunology Center, Amsterdam, The Netherlands
| | - Theo Rispens
- Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Maassen JM, van Ouwerkerk L, Allaart CF. Tapering of disease-modifying antirheumatic drugs: an overview for daily practice. THE LANCET. RHEUMATOLOGY 2021; 3:e659-e670. [PMID: 38287612 DOI: 10.1016/s2665-9913(21)00224-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/25/2021] [Accepted: 07/08/2021] [Indexed: 01/31/2024]
Abstract
In this Review, we discuss the possibility of drug tapering in patients with rheumatoid arthritis in remission or low disease activity, for glucocorticoids and disease-modifying antirheumatic drugs. We review international guidelines and recommendations, as well as remaining uncertainties, and provide an overview of the current literature. Three strategies of tapering are discussed: (1) tapering by discontinuation of one of the drugs in combination therapy regimens, (2) tapering by reducing the dose of one of the drugs in combination therapy regimens, and (3) tapering by dose reduction of monotherapy with disease-modifying antirheumatic drugs. We discuss the outcomes and robustness of evidence of trials and observational cohorts, and we give a trajectory for further research and drug tapering in daily practice.
Collapse
Affiliation(s)
| | - Lotte van Ouwerkerk
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | | |
Collapse
|
20
|
Sellam J, Morel J, Tournadre A, Bouhnik Y, Cornec D, Devauchelle-Pensec V, Dieudé P, Goupille P, Jullien D, Kluger N, Lazaro E, Le Goff B, de Lédinghen V, Lequerré T, Nocturne G, Seror R, Truchetet ME, Verhoeven F, Pham T, Richez C. PRACTICAL MANAGEMENT of patients on anti-TNF therapy: Practical guidelines drawn up by the Club Rhumatismes et Inflammation (CRI). Joint Bone Spine 2021; 88:105174. [PMID: 33992225 DOI: 10.1016/j.jbspin.2021.105174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Jérémie Sellam
- Service de Rhumatologie, CHU Saint-Antoine, Paris, France
| | - Jacques Morel
- Service de Rhumatologie, CHU Montpellier, Montpellier, France
| | - Anne Tournadre
- Service de Rhumatologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Yoram Bouhnik
- Service de Gastro-entérologie, CHU Hôpital Beaujon, Clichy, France
| | - Divi Cornec
- Service de Rhumatologie, CHRU La Cavale Blanche, Brest, France
| | | | - Philippe Dieudé
- Service de Rhumatologie, CHU Bichat-Claude Bernard, Paris, France
| | | | | | - Nicolas Kluger
- Dpt Dermatology, Helsinki, Finland; Service de Dermatologie, CHU Bichat-Claude Bernard, Paris, France
| | - Estibaliz Lazaro
- Service de Médecine interne, Hôpital Haut-Lévêque, CHU Bordeaux, Pessac, France
| | | | - Victor de Lédinghen
- Unité d'Hépatologie et transplantation hépatique, Hôpital Haut-Lévêque, CHU Bordeaux, Pessac, France
| | | | | | - Raphaèle Seror
- Service de Rhumatologie, Bicêtre, Le Kremlin-Bicêtre, France
| | | | | | - Thao Pham
- Service de Rhumatologie, CHU Sainte-Marguerite, Marseille, France
| | | |
Collapse
|
21
|
Cost-Effectiveness of Therapeutic Drug Monitoring-Guided Adalimumab Therapy in Rheumatic Diseases: A Prospective, Pragmatic Trial. Rheumatol Ther 2021; 8:1323-1339. [PMID: 34278555 PMCID: PMC8380594 DOI: 10.1007/s40744-021-00345-5] [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: 05/28/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction To assess the clinical and cost-effectiveness of therapeutic drug monitoring (TDM) based on serum adalimumab levels compared to standard of care in patients with rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis.
Methods This was a non-inferiority, multicentric, non-randomized, pragmatic trial including adult patients diagnosed with moderate-to-severe, clinically stable rheumatic diseases treated with adalimumab. Consecutive patients were assigned 1:2 to the control (CG) or the intervention group (IG), based on the site of inclusion, and followed up for 18 months. Adalimumab serum levels were measured at each study visit and released to the IG only to modify dosing strategy. Data on disease activity, healthcare resource utilization and health-related quality of life (HRQoL) measured through the EQ-5D-5L were collected. Number of persistent and overall flares, time to first flare, days experiencing high disease activity, total direct costs, quality-adjusted life years (QALYs) and incremental cost-effectiveness ratio (ICER) were calculated. Results Of the 169 recruited patients, 150 were included in the analysis (52 and 98 patients in the CG and IG, respectively). The primary endpoint was not met as persistent flares were not significantly lower in the IG, although mean (SD) number of flares was numerically lower in the IG (0.67 [0.70] versus 0.90 [0.82], P = 0.073), respectively. Based on EQ-5D-5L utilities, HRQoL was significantly higher in the IG at 3 (P = 0.001) and 6 months (P = 0.035), which overall translated into 0.075 QALYs gained per patient for the IG at month 18. Overall, direct costs were significantly lower for the IG patients (€15,311.59 [4,870.04] versus €17,378.46 [6,556.51], P = 0.030), resulting in the intervention being dominant, leading to increased QALY at a lower overall cost Conclusion Adalimumab dose tapering based on TDM for rheumatic patients led to an increased quality of life and QALY gain and entailed lower costs, being a more cost-effective alternative than clinically guided management. Supplementary Information The online version contains supplementary material available at 10.1007/s40744-021-00345-5.
Collapse
|
22
|
Jung SY, Koh JH, Kim KJ, Park YW, Yang HI, Choi SJ, Lee J, Choi CB, Kim WU. Switching from TNFα inhibitor to tacrolimus as maintenance therapy in rheumatoid arthritis after achieving low disease activity with TNFα inhibitors and methotrexate: 24-week result from a non-randomized, prospective, active-controlled trial. Arthritis Res Ther 2021; 23:182. [PMID: 34233727 PMCID: PMC8265052 DOI: 10.1186/s13075-021-02566-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022] Open
Abstract
Background Tapering or stopping biological disease-modifying anti-rheumatic drugs has been proposed for patients with rheumatoid arthritis (RA) in remission, but it frequently results in high rates of recurrence. This study evaluates the efficacy and safety of tacrolimus (TAC) as maintenance therapy in patients with established RA in remission after receiving combination therapy with tumor necrosis factor inhibitor (TNFi) and methotrexate (MTX). Methods This 24-week, prospective, open-label trial included patients who received TNFi and MTX at stable doses for ≥24 weeks and had low disease activity (LDA), measured by Disease Activity Score-28 for ≥12 weeks. Patients selected one of two arms: maintenance (TNFi plus MTX) or switched (TAC plus MTX). The primary outcome was the difference in the proportion of patients maintaining LDA at week 24, which was assessed using a logistic regression model. Adverse events were monitored throughout the study period. Results In efficacy analysis, 80 and 34 patients were included in the maintenance and switched arms, respectively. At week 24, LDA was maintained in 99% and 91% of patients in the maintenance and switched arms, respectively (odds ratio, 0.14; 95% confidence interval, 0.01–1.59). Drug-related adverse effects tended to be more common in the switched arm than in the maintenance arm (20.9% versus 7.1%, respectively) but were well-tolerated. Conclusion This controlled study tested a novel treatment strategy of switching from TNFi to TAC in RA patients with sustained LDA, and the findings suggested that TNFi can be replaced with TAC in most patients without the patients experiencing flare-ups for at least 24 weeks. Trial registration Korea CDC CRIS, KCT0005868. Registered 4 February 2021—retrospectively registered Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02566-z.
Collapse
Affiliation(s)
- Sang Youn Jung
- Division of Rheumatology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jung Hee Koh
- Division of Rheumatology, Department of Internal Medicine, Bucheon St. Mary's Hospital, the Catholic University of Korea, Seoul, South Korea
| | - Ki-Jo Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent Hospital, the Catholic University of Korea, Seoul, South Korea
| | - Yong-Wook Park
- Division of Rheumatology, Department of Internal Medicine, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Hyung-In Yang
- College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Sung Jae Choi
- Division of Rheumatology, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, South Korea
| | - Jisoo Lee
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Chan-Bum Choi
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St Mary's Hospital, Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, the Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.
| |
Collapse
|
23
|
Martínez-Feito A, Navarro-Compán V, Hernández-Breijo B, Olariaga-Mérida E, Peiteado D, Villalba A, Nuño L, Monjo I, Diego C, Pascual-Salcedo D, Nozal P, Balsa A, Plasencia-Rodríguez C. Early monitoring of infliximab serum trough levels predicts long-term therapy failure in patients with axial spondyloarthritis. Scand J Rheumatol 2021; 51:102-109. [PMID: 34182885 DOI: 10.1080/03009742.2021.1914430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Objective: To evaluate whether serum infliximab trough levels (ITL) during the early stages of treatment are predictive of long-term clinical failure in patients with axial spondyloarthritis (axSpA).Method: Longitudinal observational study involving 81 patients with axSpA monitored during infliximab therapy. Serum ITL were measured before starting infliximab treatment and at weeks 2 (W2), W6 and W12 of treatment. Disease activity was assessed by Ankylosing Spondylitis Disease Activity Score (ASDAS) at baseline, W24 and W52, and every 6 months thereafter until treatment discontinuation, regardless of the reason. Non-clinically important improvement was defined by ΔASDAS<1.1. The association between serum levels during the early stages and clinical outcomes (non-clinically important improvement at W52, drug survival and drop-out due to secondary inefficacy) was investigated through logistic regression models and Kaplan Meier curves. Receiver operating characteristic (ROC) curves were employed to determine the best cut-off for serum ITL.Results: Out of the 81 patients, 45 (56%) did not achieve clinical improvement at W52. These patients had lower serum ITL at W12 compared to those who improved: ITL [median (IQR)]: 4.1(0.9-8.3) µg/mL vs 7.1 (4.3-11.3) µg/mL, respectively;p = 0.007). ITL<6.7 µg/mL at W12 was significantly associated with: i) not achieving clinical improvement at W52 (OR: 2.3; 95%CI: 1.3-3.9); ii) shorter drug survival (5.0 years (95% CI 3.8-6.2) vs 7.0 years (95% CI 4.8-6.9; p = 0.04), and iii) higher drop-out rates due to secondary inefficacy (OR: 3.5; 95% CI: 1.2-10.2).Conclusion: Low serum ITL at W12 were associated with long-term clinical failure in patients with axSpA, due to secondary inefficacy.
Collapse
Affiliation(s)
- A Martínez-Feito
- Immunology Unit, La Paz University Hospital, Madrid, Spain.,Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain
| | - V Navarro-Compán
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain.,Rheumatology Department, La Paz University Hospital, Madrid, Spain
| | - B Hernández-Breijo
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain
| | - E Olariaga-Mérida
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain
| | - D Peiteado
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain.,Rheumatology Department, La Paz University Hospital, Madrid, Spain
| | - A Villalba
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain.,Rheumatology Department, La Paz University Hospital, Madrid, Spain
| | - L Nuño
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain.,Rheumatology Department, La Paz University Hospital, Madrid, Spain
| | - I Monjo
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain.,Rheumatology Department, La Paz University Hospital, Madrid, Spain
| | - C Diego
- Immunology Unit, La Paz University Hospital, Madrid, Spain
| | - D Pascual-Salcedo
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain
| | - P Nozal
- Immunology Unit, La Paz University Hospital, Madrid, Spain
| | - A Balsa
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain.,Rheumatology Department, La Paz University Hospital, Madrid, Spain
| | - C Plasencia-Rodríguez
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research, Madrid, Spain.,Rheumatology Department, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
24
|
Nassar-Sheikh Rashid A, Schonenberg-Meinema D, Bergkamp SC, Bakhlakh S, de Vries A, Rispens T, Kuijpers TW, Wolbink G, van den Berg JM. Therapeutic drug monitoring of anti-TNF drugs: an overview of applicability in daily clinical practice in the era of treatment with biologics in juvenile idiopathic arthritis (JIA). Pediatr Rheumatol Online J 2021; 19:59. [PMID: 33926495 PMCID: PMC8082819 DOI: 10.1186/s12969-021-00545-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/14/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Anti-tumor necrosis factor (TNF) drugs have improved the prognosis for juvenile idiopathic arthritis (JIA) significantly. However, evidence for individual treatment decisions based on serum anti-TNF drug levels and the presence of anti-drug antibodies (ADAbs) in children is scarce. We aimed to assess if anti-TNF drug levels and/or ADAbs influenced physician's treatment decisions in children with JIA. METHODS Patients' records in our center were retrospectively screened for measurements of anti-TNF drug levels and ADAbs in children with JIA using etanercept, adalimumab or infliximab. Clinical characteristics and disease activity were retrieved from patient charts. RESULTS We analyzed 142 measurements of anti-TNF drug levels in 65 children with JIA. Of these, ninety-seven (68.3%) were trough concentrations. N = 14/97 (14.4%) of these showed trough concentrations within the therapeutic drug range known for adults with RA and IBD. ADAbs against adalimumab were detected in seven patients and against infliximab in one patient. Seven (87,5%) of these ADAb-positive patients had non-detectable drug levels. A flowchart was made on decisions including rational dose escalation, stopping treatment in the presence of ADAbs and undetectable drug levels, showing that 45% of measurements influenced treatment decisions, which concerned 65% of patients (n = 42/65). CONCLUSIONS In the majority of patients, measurement of anti-TNF drug levels led to changes in treatment. A wide variation of anti-TNF drug levels was found possibly due to differences in drug clearance in different age groups. There is need for determination of therapeutic drug ranges and pharmacokinetic curves for anti-TNF and other biologics in children with JIA.
Collapse
Affiliation(s)
- A. Nassar-Sheikh Rashid
- grid.7177.60000000084992262Zaans Medical Center, Zaandam and Emma Children’s Hospital, Amsterdam UMC, Pediatric Immunology, Rheumatology and Infectious Diseases, University of Amsterdam, Amsterdam, The Netherlands
| | - D. Schonenberg-Meinema
- grid.7177.60000000084992262Emma Children’s Hospital, Amsterdam UMC, Pediatric Immunology, Rheumatology and Infectious Diseases, University of Amsterdam, Amsterdam, The Netherlands
| | - S. C. Bergkamp
- grid.7177.60000000084992262Emma Children’s Hospital, Amsterdam UMC, Pediatric Immunology, Rheumatology and Infectious Diseases, University of Amsterdam, Amsterdam, The Netherlands
| | - S. Bakhlakh
- grid.7177.60000000084992262Emma Children’s Hospital, Amsterdam UMC, Pediatric Immunology, Rheumatology and Infectious Diseases, University of Amsterdam, Amsterdam, The Netherlands
| | - A. de Vries
- grid.417732.40000 0001 2234 6887Department of Immunopathology, Sanquin Diagnostic Services Amsterdam, Amsterdam, The Netherlands
| | - T. Rispens
- grid.417732.40000 0001 2234 6887Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - T. W. Kuijpers
- grid.7177.60000000084992262Emma Children’s Hospital, Amsterdam UMC, Pediatric Immunology, Rheumatology and Infectious Diseases, University of Amsterdam, Amsterdam, The Netherlands
| | - G. Wolbink
- grid.418029.60000 0004 0624 3484Department of Immunopathology, CLB Sanquin Amsterdam and Department of Rheumatology, Jan van Breemen Institute Amsterdam, Amsterdam, The Netherlands
| | - J. M. van den Berg
- grid.7177.60000000084992262Emma Children’s Hospital, Amsterdam UMC, Pediatric Immunology, Rheumatology and Infectious Diseases, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Wadhwa M, Bird C, Atkinson E, Cludts I, Rigsby P. The First WHO International Standard for Adalimumab: Dual Role in Bioactivity and Therapeutic Drug Monitoring. Front Immunol 2021; 12:636420. [PMID: 33936049 PMCID: PMC8082443 DOI: 10.3389/fimmu.2021.636420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
The expanded availability of adalimumab products continues to widen patient access and reduce costs with substantial benefit to healthcare systems. However, the long-term success of these medicines is highly dependent on maintaining consistency in quality, safety and efficacy while minimizing any risk of divergence during life-cycle management. In recognition of this need and demand from global manufacturers, the World Health Organization (WHO) Expert Committee on Biological standardization established the WHO 1st International standard (IS) for Adalimumab (coded 17/236) in October 2019 with a defined unitage ascribed to each of the individual bioactivities evaluated in the study namely, TNF-α binding, TNF-α neutralization, complement dependent cytotoxicity and antibody-dependent cellular cytotoxicity. For development of the IS, two candidate standards were manufactured as per WHO recommendations. Analysis of extensive datasets generated by testing of a common set of samples including the candidate standards by multiple stakeholders including regulatory agencies using their own qualified assays in a large international collaborative study showed comparable biological activity for the tested candidates for the different activities. Use of a common standard significantly decreased the variability of bioassays and improved agreement in potency estimates. Data from this study clearly supports the utility of the IS as an important tool for assuring analytical assay performance, for bioassay calibration and validation, for identifying and controlling changes in bioactivity during life-cycle management and for global harmonization of adalimumab products. In addition, in a separate multi-center study which included involvement of hospital and clinical diagnostic laboratories, the suitability of the adalimumab IS for therapeutic drug monitoring assays was examined by analysis of data from testing of a common blind coded panel of adalimumab spiked serum samples representative of the clinical scenario along with the IS and in-house standards in diverse immunoassays/platforms. Both commercially available and in-house assays that are routinely used for assessing adalimumab trough levels were included. Excellent agreement in estimates for adalimumab content in the spiked samples was observed regardless of the standard or the method with inter-laboratory variability also similar regardless of the standard employed. This data, for the first time, provides support for the extended applicability of the IS in assays in use for therapeutic drug monitoring based on the mass content of the IS. The adalimumab IS, in fulfilling clinical demand, can help toward standardizing and harmonizing clinical monitoring assays for informed clinical decisions and/or personalized treatment strategies for better patient outcomes. Collectively, a significant role for the adalimumab IS in assuring the quality, safety and efficacy of adalimumab products globally is envisaged.
Collapse
Affiliation(s)
- Meenu Wadhwa
- Biotherapeutics Division, National Institute for Biological Standards and Control, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Chris Bird
- Biotherapeutics Division, National Institute for Biological Standards and Control, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Eleanor Atkinson
- Analytical and Biological Sciences Division, National Institute for Biological Standards and Control, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Isabelle Cludts
- Biotherapeutics Division, National Institute for Biological Standards and Control, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Peter Rigsby
- Analytical and Biological Sciences Division, National Institute for Biological Standards and Control, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| |
Collapse
|
26
|
Tikhonova IA, Yang H, Bello S, Salmon A, Robinson S, Hemami MR, Dodman S, Kharechko A, Haigh RC, Jani M, McDonald TJ, Hoyle M. Enzyme-linked immunosorbent assays for monitoring TNF-alpha inhibitors and antibody levels in people with rheumatoid arthritis: a systematic review and economic evaluation. Health Technol Assess 2021; 25:1-248. [PMID: 33555998 PMCID: PMC7898084 DOI: 10.3310/hta25080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis is a chronic autoimmune disease that primarily causes inflammation, pain and stiffness in the joints. People with severe disease may be treated with biological disease-modifying anti-rheumatic drugs, including tumour necrosis factor-α inhibitors, but the efficacy of these drugs is hampered by the presence of anti-drug antibodies. Monitoring the response to these treatments typically involves clinical assessment using response criteria, such as Disease Activity Score in 28 joints or European League Against Rheumatism. Enzyme-linked immunosorbent assays can also be used to measure drug and antibody levels in the blood. These tests may inform whether or not adjustments to treatment are required or help clinicians to understand the reasons for treatment non-response or a loss of response. METHODS Systematic reviews were conducted to identify studies reporting on the clinical effectiveness and cost-effectiveness of using enzyme-linked immunosorbent assays to measure drug and anti-drug antibody levels to monitor the response to tumour necrosis factor-α inhibitors [adalimumab (Humira®; AbbVie, Inc., North Chicago, IL, USA), etanercept (Enbrel®; Pfizer, Inc., New York, NY, USA), infliximab (Remicade®, Merck Sharp & Dohme Limited, Hoddesdon, UK), certolizumab pegol (Cimzia®; UCB Pharma Limited, Slough, UK) and golimumab (Simponi®; Merck Sharp & Dohme Limited)] in people with rheumatoid arthritis who had either achieved treatment target (remission or low disease activity) or shown primary or secondary non-response to treatment. A range of bibliographic databases, including MEDLINE, EMBASE and CENTRAL (Cochrane Central Register of Controlled Trials), were searched from inception to November 2018. The risk of bias was assessed using the Cochrane ROBINS-1 (Risk Of Bias In Non-randomised Studies - of Interventions) tool for non-randomised studies, with adaptations as appropriate. Threshold and cost-utility analyses that were based on a decision tree model were conducted to estimate the economic outcomes of adding therapeutic drug monitoring to standard care. The costs and resource use were considered from the perspective of the NHS and Personal Social Services. No discounting was applied to the costs and effects owing to the short-term time horizon of 18 months that was adopted in the economic analysis. The impact on the results of variations in testing and treatment strategies was explored in numerous clinically plausible sensitivity analyses. RESULTS Two studies were identified: (1) a non-randomised controlled trial, INGEBIO, that compared standard care with therapeutic drug monitoring using Promonitor® assays [Progenika Biopharma SA (a Grifols-Progenika company), Derio, Spain] in Spanish patients receiving adalimumab who had achieved remission or low disease activity; and (2) a historical control study. The economic analyses were informed by INGEBIO. Different outcomes from INGEBIO produced inconsistent results in both threshold and cost-utility analyses. The cost-effectiveness of therapeutic drug monitoring varied, from the intervention being dominant to the incremental cost-effectiveness ratio of £164,009 per quality-adjusted life-year gained. However, when the frequency of testing was assumed to be once per year and the cost of phlebotomy appointments was excluded, therapeutic drug monitoring dominated standard care. LIMITATIONS There is limited relevant research evidence and much uncertainty about the clinical effectiveness and cost-effectiveness of using enzyme-linked immunosorbent assay-based testing for therapeutic drug monitoring in rheumatoid arthritis patients. INGEBIO had serious limitations in relation to the National Institute for Health and Care Excellence scope: only one-third of participants had rheumatoid arthritis, the analyses were mostly not by intention to treat and the follow-up was 18 months only. Moreover, the outcomes might not be generalisable to the NHS. CONCLUSIONS Based on the available evidence, no firm conclusions could be made about the cost-effectiveness of therapeutic drug monitoring in England and Wales. FUTURE WORK Further controlled trials are required to assess the impact of using enzyme-linked immunosorbent assays for monitoring the anti-tumour necrosis factors in people with rheumatoid arthritis. STUDY REGISTRATION This study is registered as PROSPERO CRD42018105195. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 25, No. 8. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Irina A Tikhonova
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
- Southampton Health Technology Assessments Centre, University of Southampton, Southampton, UK
| | - Huiqin Yang
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Segun Bello
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Andrew Salmon
- Peninsula Collaboration for Health Operational Research and Development, University of Exeter Medical School, Exeter, UK
| | - Sophie Robinson
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Mohsen Rezaei Hemami
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Sophie Dodman
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Andriy Kharechko
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | | | - Meghna Jani
- Division of Musculoskeletal & Dermatological Sciences, University of Manchester, Manchester, UK
| | - Timothy J McDonald
- Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
- University of Exeter Medical School, Exeter, UK
| | - Martin Hoyle
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| |
Collapse
|
27
|
Strand V, Goncalves J, Isaacs JD. Immunogenicity of biologic agents in rheumatology. Nat Rev Rheumatol 2020; 17:81-97. [PMID: 33318665 DOI: 10.1038/s41584-020-00540-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Biologic agents have become a core component of therapeutic strategies for many inflammatory rheumatic diseases. However, perhaps reflecting the specificity and generally high affinity of biologic agents, these therapeutics have been used by rheumatologists with less consideration of their pharmacokinetics than that of conventional synthetic DMARDs. Immunogenicity was recognized as a potential limitation to the use of biologic agents at an early stage in their development, although regulatory guidance was relatively limited and assays to measure immunogenicity were less sophisticated than today. The advent of biosimilars has sparked a renewed interest in immunogenicity that has resulted in the development of increasingly sensitive assays, an enhanced appreciation of the pharmacokinetic consequences of immunogenicity and the development of comprehensive and specific guidance from regulatory authorities. As a result, rheumatologists have a greatly improved understanding of the field in general, including the factors responsible for immunogenicity, its potential clinical consequences and the implications for everyday treatment. In some specialties, immunogenicity testing is becoming a part of routine clinical management, but definitive evidence of its cost-effectiveness in rheumatology is awaited.
Collapse
Affiliation(s)
- Vibeke Strand
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, CA, USA.
| | - Joao Goncalves
- Research Institute for Medicines (iMed), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
28
|
Reduction of biologics in rheumatoid arthritis: a systematic review and meta-analysis. Rheumatol Int 2020; 40:1949-1959. [PMID: 32710197 DOI: 10.1007/s00296-020-04651-z] [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: 03/26/2020] [Accepted: 07/11/2020] [Indexed: 10/23/2022]
Abstract
The effects of dose reduction or spacing of all types of biologics in rheumatoid arthritis has not been consistently assessed in systematic reviews. We aimed to assess the effects of biologics reduction compared with dose maintenance in patients with rheumatoid arthritis in low disease activity or remission. We performed a systematic review with meta-analysis according to a previously registered protocol (PROSPERO registration: CRD42017069080); and searched MEDLINE, Embase, Scopus, Cochrane Library and trial registers up to July, 2020. Two researchers selected, extracted and assessed the risk of bias of controlled trials that randomized patients to reduction/spacing or dose maintenance of biologics. Low disease activity, disability and other clinically important outcomes were summarized in random effect meta-analyses. We rated the certainty of evidence according to the Grading of Recommendations Assessment, Development, and Evaluation approach. We included ten studies (n = 1331 patients), which assessed reduction or spacing of abatacept, adalimumab, certolizumab pegol, etanercept, or tocilizumab. Risk of bias was high in over half of trials, mainly due to lack of blinding. No statistically significant difference was found in low disease activity (RR = 0.90; 95% CI 0.78-1.04; I2 = 60%, very low certainty), and other outcomes. Subgroup analysis of blinded studies led to homogeneous results, which remained heterogeneous in open-label studies. Reduction or spacing biologics did not affect disease activity and other important outcome. Changes in the doses regimen should consider patient preferences, considering the low certainty of evidence.
Collapse
|
29
|
Pedersen L, Szecsi PB, Johansen PB, Bjerrum PJ. Evaluation of Therapeutic Drug Monitoring in the Clinical Management of Patients with Rheumatic Diseases: Data from a Retrospective Single-Center Cohort Study. Biologics 2020; 14:115-125. [PMID: 33162753 PMCID: PMC7643816 DOI: 10.2147/btt.s262511] [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: 05/12/2020] [Accepted: 07/24/2020] [Indexed: 01/13/2023]
Abstract
Purpose Treatment of rheumatic diseases with tumor necrosis factor inhibitors leads to improved clinical outcomes. Therapeutic drug monitoring (TDM) may assist in guiding clinical decisions. This study investigates the impact of TDM on clinical outcome, decision-making and biologics cost expenditure. Patients and Methods In a retrospective observational study of 306 patients with rheumatic diseases treated with four different tumor necrosis factor inhibitors, drug levels and antidrug antibodies were measured over a period of one year. Primary outcomes were the clinicians’ response to each TDM result and the clinical outcome two years after TDM initiation. Outcomes were compared between the 111 TDM-guided patients and the 195 empirically guided patients. Results Treatment change occurred in 55% of the patients in the TDM group, but in only 38% in the empirically guided group. In the TDM group, 89 (79.5%) patients were in remission or had low disease activity after two years follow-up compared to 128 (65.6%) patients in the empirical group. The average cost of biologics per patient per year was lower in the TDM group than in the empirical group for patients receiving infliximab, adalimumab or etanercept at baseline but not for golimumab. Conclusion TDM-guided decision-making is useful in rheumatic patients receiving TNFi and may optimize therapeutic decisions, leading to a better control of disease activity. Proactive TDM may support decisions on dose tapering, resulting in lower drug consumption and biologics cost expenditure.
Collapse
Affiliation(s)
- Lise Pedersen
- Department of Clinical Biochemistry, University of Copenhagen, Holbæk Hospital, Holbæk, Denmark
| | - Pal Bela Szecsi
- Department of Clinical Biochemistry, University of Copenhagen, Holbæk Hospital, Holbæk, Denmark
| | - Per Birger Johansen
- Department of Rheumatology, University of Copenhagen, Holbæk Hospital, Holbæk, Denmark
| | - Poul Jannik Bjerrum
- Department of Clinical Biochemistry, University of Copenhagen, Holbæk Hospital, Holbæk, Denmark
| |
Collapse
|
30
|
Marotte H, Rinaudo-Gaujous M, Petiet C, Fautrel B, Paul S. Tapering without relapse in rheumatoid arthritis patients with high TNF blocker concentrations: data from STRASS study. Ann Rheum Dis 2020; 79:e81. [PMID: 31048287 DOI: 10.1136/annrheumdis-2019-215546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 04/17/2019] [Indexed: 11/04/2022]
Affiliation(s)
- Hubert Marotte
- Rhumatologie, CHU Saint-Etienne, Saint-Etienne, France
- SAINBIOSE, INSERM U1059, University of Lyon, Saint-Etienne, France
| | | | - Cécile Petiet
- Rhumatologie, CHU Saint-Etienne, Saint-Etienne, France
| | - Bruno Fautrel
- Rheumatology, Assistance Publique-Hopitaux de Paris, Paris, France
- GRC08-IPLESP, UPMC Faculte de Medecine, Paris, France
| | - Stéphane Paul
- Laboratory of Immunology and Immunomonitoring, Universite Jean Monnet Saint-Etienne Faculte de Medecine Jacques Lisfranc, Saint Priest en Jarez, France
| |
Collapse
|
31
|
l'Ami MJ, Krieckaert CL, Nurmohamed MT, van Vollenhoven RF, Rispens T, Boers M, Wolbink GJ. Response to: 'Tapering without relapse in rheumatoid arthritis patients with high TNF blocker concentrations: data from the STRASS study' by Marotte et al. Ann Rheum Dis 2020; 79:e82. [PMID: 31072816 DOI: 10.1136/annrheumdis-2019-215609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/03/2019] [Indexed: 11/03/2022]
Affiliation(s)
- Merel J l'Ami
- Rheumatology, Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
| | - Charlotte L Krieckaert
- Rheumatology, Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
| | - Michael T Nurmohamed
- Rheumatology, Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center | Amsterdam Medical Center, VU Medical Center, Amsterdam, The Netherlands
| | - Ronald F van Vollenhoven
- Amsterdam Rheumatology and Immunology Center | Amsterdam Medical Center, VU Medical Center, Amsterdam, The Netherlands
| | - Theo Rispens
- Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Maarten Boers
- Amsterdam Rheumatology and Immunology Center | Amsterdam Medical Center, VU Medical Center, Amsterdam, The Netherlands
- Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Gerrit Jan Wolbink
- Rheumatology, Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
- Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Lorenzin M, Ometto F, Ortolan A, Felicetti M, Favero M, Doria A, Ramonda R. An update on serum biomarkers to assess axial spondyloarthritis and to guide treatment decision. Ther Adv Musculoskelet Dis 2020; 12:1759720X20934277. [PMID: 32636944 PMCID: PMC7315656 DOI: 10.1177/1759720x20934277] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022] Open
Abstract
Axial spondyloarthritis (axSpA) is a group of debilitating, chronic, rheumatic conditions characterized by inflammation and new bone formation, mainly involving the spine and the sacroiliac joints. The lack of biomarkers in axSpA is well known. Despite significant treatment advances in recent years thanks to the introduction of drugs with a new mode of action, such as new biologic and targeted synthetic disease-modifying antirheumatic drugs, no relevant improvement in the identification of disease biomarkers has been achieved. Common parameters, such as erythrocyte sedimentation rate and C-reactive protein, which are routinely used to measure systemic inflammation, are the sole markers available to date and are not adequate to assess disease activity in all patients. The aim of this study is to review the most promising serum biomarkers that may help treatment decision in axSpA via a proper assessment of disease activity and identification of negative prognostic factors.
Collapse
Affiliation(s)
- Mariagrazia Lorenzin
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Francesca Ometto
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Augusta Ortolan
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Mara Felicetti
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Via Giustiniani 2, Padova, 35128, Italy
| |
Collapse
|
33
|
Evaluation of dose-tapering strategies for intravenous tocilizumab in rheumatoid arthritis patients using model-based pharmacokinetic/pharmacodynamic simulations. Eur J Clin Pharmacol 2020; 76:1417-1425. [PMID: 32514745 DOI: 10.1007/s00228-020-02925-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Tocilizumab is a humanized monoclonal antibody approved for rheumatoid arthritis treatment. In clinical practice, empirical dose-tapering strategies are implemented in patients showing sustained remission or low disease activity (LDA) to avoid overtreatment and reduce costs. Since rational adaptive-dosing algorithms taking the full pharmacokinetic (PK)/pharmacodynamic (PD) characteristics into account are currently lacking, we aimed to develop novel tapering strategies and compare them with currently used empirical ones. METHODS Four strategies were simulated on a virtual population. In all of them, the same initial dose was administered every 28 days for six consecutive months. Then, different strategies were considered: (1) label-dosing; (2) mild empirical dose-tapering; (3) intense empirical dose-tapering; (4) therapeutic drug monitoring (TDM)-guided dose-tapering. The different strategies were evaluated on the proportion of patients who maintain remission/LDA 1 year after the intervention. Cost-savings of direct drug costs were also estimated as relative dose intensity. RESULTS The overall proportion of simulated patients in remission/LDA after 1 year of the intervention was comparable between the mild empirical and the TDM-guided dose-tapering strategies, and much lower for the intense empirical dose-tapering strategy (80.3%, 78.2%, and 69.0%, respectively). Likewise, 1-year flare rates were lower for the mild empirical and TDM-guided tapering strategies. The relative dose intensity was lowest for the intense empirical dose-tapering, followed by the TDM-guided and the mild empirical dose-tapering approaches (61.2%, 71.0%, and 80.4%, respectively). CONCLUSION We demonstrated that the TDM-guided strategy using model-based algorithms performed similarly to mild empirical dose-tapering strategies in overall remission/LDA rates but is superior in cost-savings.
Collapse
|
34
|
Kerschbaumer A, Sepriano A, Smolen JS, van der Heijde D, Dougados M, van Vollenhoven R, McInnes IB, Bijlsma JWJ, Burmester GR, de Wit M, Falzon L, Landewé R. Efficacy of pharmacological treatment in rheumatoid arthritis: a systematic literature research informing the 2019 update of the EULAR recommendations for management of rheumatoid arthritis. Ann Rheum Dis 2020; 79:744-759. [PMID: 32033937 PMCID: PMC7286044 DOI: 10.1136/annrheumdis-2019-216656] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To inform the 2019 update of the European League against Rheumatism (EULAR) recommendations for the management of rheumatoid arthritis (RA). METHODS A systematic literature research (SLR) to investigate the efficacy of any disease-modifying antirheumatic drug (DMARD) (conventional synthetic (cs)DMARD, biological (b) and biosimilar DMARD, targeted synthetic (ts)DMARD) or glucocorticoid (GC) therapy in patients with RA was done by searching MEDLINE, Embase and the Cochrane Library for articles published between 2016 and 8 March 2019. RESULTS 234 abstracts were selected for detailed assessment, with 136 finally included. They comprised the efficacy of bDMARDs versus placebo or other bDMARDs, efficacy of Janus kinase (JAK) inhibitors (JAKi) across different patient populations and head-to-head of different bDMARDs versus JAKi or other bDMARDs. Switching of bDMARDs to other bDMARDs or tsDMARDs, strategic trials and tapering studies of bDMARDs, csDMARDs and JAKi were assessed. The drugs evaluated included abatacept, adalimumab, ABT-122, baricitinib, certolizumab pegol, SBI-087, CNTO6785, decernotinib, etanercept, filgotinib, golimumab, GCs, GS-9876, guselkumab, hydroxychloroquine, infliximab, leflunomide, mavrilimumab, methotrexate, olokizumab, otilimab, peficitinib, rituximab, sarilumab, salazopyrine, secukinumab, sirukumab, tacrolimus, tocilizumab, tofacitinib, tregalizumab, upadacitinib, ustekinumab and vobarilizumab. The efficacy of many bDMARDs and tsDMARDs was shown. Switching to another tumour necrosis factor inhibitor (TNFi) or non-TNFi bDMARDs after TNFi treatment failure is efficacious. Tapering of DMARDs is possible in patients achieving long-standing stringent clinical remission; in patients with residual disease activity (including patients in LDA) the risk of flares is increased during the tapering. Biosimilars are non-inferior to their reference products. CONCLUSION This SLR informed the task force regarding the evidence base of various therapeutic regimen for the development of the update of EULAR's RA management recommendation.
Collapse
Affiliation(s)
| | - Alexandre Sepriano
- Leiden University Medical Center, Leiden, The Netherlands
- NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | | | | | | | | | | | | | | | | | | | - Robert Landewé
- Amsterdam Rheumatology Center, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Vinson D, Molet-Benhamou L, Degboé Y, den Broeder A, Ibrahim F, Pontes C, Westhovens R, Závada J, Pham T, Barnetche T, Constantin A, Ruyssen-Witrand A. Impact of tapering targeted therapies (bDMARDs or JAKis) on the risk of serious infections and adverse events of special interest in patients with rheumatoid arthritis or spondyloarthritis: a systematic analysis of the literature and meta-analysis. Arthritis Res Ther 2020; 22:97. [PMID: 32349791 PMCID: PMC7191828 DOI: 10.1186/s13075-020-02188-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES To systematically review the impact of tapering targeted therapies (bDMARDs or JAKis) on the risk of serious infections and severe adverse events (SAEs) in patients with rheumatoid arthritis (RA) or axial spondyloarthritis (axSpA) in remission or low disease activity (LDA) state. MATERIALS AND METHODS A meta-analysis based on a systematic review of PubMed, Embase, Cochrane, until August 2019, as well as relevant databases of international conferences, was used to evaluate the risk difference (RD) at 95% confidence interval (95% CI) of incidence density of serious infections, SAEs, malignancies, cardiovascular adverse events (CV AEs), or deaths after tapering (dose reduction or spacing) compared to continuation of targeted therapies. RESULTS Of the 1957 studies initially identified, 13 controlled trials (9 RA and 4 SpA trials) were included in the meta-analysis. 1174 patient-years were studied in the tapering group (TG) versus 1086 in the usual care group (UC). There were 1.7/100 patient-year (p-y) serious infections in TG versus 2.6/100 p-y in UC (RD (95% CI) 0.01 (0.00 to 0.02), p = 0.13) and 7.4/100 p-y SAEs in TG versus 6.7/100 p-y in UC (RD 0.00 (- 0.02 to 0.02), p = 0.82). The risk of malignancies, CV AEs, or deaths did not differ between the tapering and the usual care groups. Subgroup analysis (RA and SpA) detected no significant differences between the two groups. CONCLUSION We could not show significant impact of tapering bDMARD or JAKi over continuation concerning the risk of serious infections, SAEs, malignancies, CV AEs, or deaths in RA and SpA patients in remission or LDA state.
Collapse
Affiliation(s)
- D Vinson
- Rheumatology Department, CHU Sainte Marguerite, 270 Boulevard de Sainte-Marguerite, 13009, Marseille, France.
| | | | - Y Degboé
- INSERM U1043, CPTP, Toulouse, France
| | - A den Broeder
- Department of Rheumatology, Sint Maartenskliniek, 6500 GM, Nijmegen, The Netherlands
| | - F Ibrahim
- Centre for Rheumatic Diseases, Department of Inflammation Biology, Faculty of Life Sciences and Medicine, School of Immunology and Microbial Sciences, King's College London, Cutcombe Road, London, SE5 9RJ, UK
| | - C Pontes
- Department of Pharmacology, Therapeutics and Toxicology, Unitat docent Parc Taulí-Medical School-Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - R Westhovens
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Rheumatology University Hospitals Leuven, KULeuven, Leuven, Belgium
| | - J Závada
- First Faculty of Medicine, Institute of Rheumatology and Department of Rheumatology, Charles University, Na Slupi 4, Prague, Czech Republic
| | - T Pham
- Rheumatology Department, CHU Sainte Marguerite, 270 Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - T Barnetche
- Rheumatology Department, Pellegrin Hospital, Bordeaux, France
| | - A Constantin
- Rheumatology Department, Hôpital Purpan, Toulouse, France
| | | |
Collapse
|
36
|
Factors Influencing Drug Disposition of Monoclonal Antibodies in Inflammatory Bowel Disease: Implications for Personalized Medicine. BioDrugs 2020; 33:453-468. [PMID: 31301024 DOI: 10.1007/s40259-019-00366-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Monoclonal antibody (mAb) therapies have revolutionized the treatment of several chronic inflammatory diseases, including the inflammatory bowel diseases (IBD), Crohn's disease, and ulcerative colitis. While efficacious, responses to these therapies vary considerably from patient to patient, due in part to inter- and intra-individual variability in pharmacokinetics (PK) and drug exposure. The concept of personalized medicine to monitor drug exposure and to adjust dosing in individual patients is consequently gaining acceptance as a powerful tool to optimize mAb therapy for improved outcomes in IBD. This review provides a brief overview of the different mAbs currently approved or in development for the treatment of IBD, including their presumed mechanisms of action and PK properties. Specifically described are (1) the factors known to affect mAb PK and drug exposure in patients with IBD, (2) the value of population PK/pharmacodynamic (PD) modeling to identify and understand the influence of these factors on drug exposure and effect, and (3) the clinical evidence for the potential of therapeutic drug monitoring (TDM) to improve IBD outcomes in response to mAb-based therapy. Incorporation of PK/PD parameters into clinical decision support tools has the potential to guide therapeutic decision making and aid implementation of personalized medicine strategies in patients with IBD.
Collapse
|
37
|
Perry M, Abdullah A, Frleta M, MacDonald J, McGucken A. The potential value of blood monitoring of biologic drugs used in the treatment of rheumatoid arthritis. Ther Adv Musculoskelet Dis 2020; 12:1759720X20904850. [PMID: 32095163 PMCID: PMC7011331 DOI: 10.1177/1759720x20904850] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022] Open
Abstract
The advent of biological therapies has been a major therapeutic advance in rheumatology. Many patients now enjoy improved quality of life through better disease control. The number of therapies continues to grow both within drug class (including biosimilar drugs) and via new mechanisms. For the first time, nonbiological drugs such as small-molecule inhibitors (Janus kinase inhibitors) have shown clinical equivalence. However, clinical unmet need remains with up to a third of patients commenced on a biologic therapy having minimal or no response: (a) Generally, the first biologic used secures the best response, with likelihood of remission falling thereafter with successive therapies; (b) the success of strategy trials using biological therapies can be difficult to replicate in clinical practice due to a combination of patient factors and service limitations. Accordingly, ensuring optimization of initial treatment is an important consideration before switching to alternatives. Therapeutic drug monitoring (TDM) is the measurement of serum levels of a biologic drug with the aim of improving patient care. It is usually combined with detection of any antidrug antibodies that could neutralize the effect of the therapy. This technology has the potential to be a form of 'personalized medicine' by individualizing therapy, in particular, dosing and likelihood of sustained treatment response. It requires a clear relationship between drug dose, blood concentration and therapeutic effect. This paper will outline the technology behind TDM and unpack what we can learn from our colleagues in gastroenterology, where the adoption of TDM is at a more advanced stage than in rheumatology. It will explore and set out a number of clinical scenarios where rheumatologists might find TDM helpful in day-to-day practice. Finally, an outline is given of international developments, including regulatory body appraisals and guideline development.
Collapse
Affiliation(s)
- Martin Perry
- Department Rheumatology, Royal Alexandra
Hospital, 9 Corsebar Road, Paisley Renfrewshire PA2 9PN, UK
| | - Azhar Abdullah
- Department Rheumatology, Royal Alexandra
Hospital, Paisley, UK
| | - Marina Frleta
- Department Rheumatology, Royal Alexandra
Hospital, Paisley, UK
| | - Jonathan MacDonald
- Department Gastroenterology, Queen Elizabeth
University Hospital, Glasgow, UK
| | - Andrew McGucken
- Department Rheumatology, Royal Alexandra
Hospital, Paisley, UK
| |
Collapse
|
38
|
Ding X, Zhu R, Wu J, Xue L, Gu M, Miao L. Early Adalimumab and Anti-Adalimumab Antibody Levels for Prediction of Primary Nonresponse in Ankylosing Spondylitis Patients. Clin Transl Sci 2020; 13:547-554. [PMID: 31961477 PMCID: PMC7214645 DOI: 10.1111/cts.12738] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/02/2019] [Indexed: 12/11/2022] Open
Abstract
This study aimed at exploring the concentration‐effect relationship of adalimumab and early adalimumab and anti‐adalimumab antibody (AAA) levels in predicting primary nonresponse in a real‐world pilot cohort of patients with ankylosing spondylitis. Thirty‐one patients were included. The Ankylosing Spondylitis Disease Activity Score improved with increasing adalimumab trough level at week 12 and reached a major improvement with levels between 8 and 12 μg/mL. Moreover, weeks 4 and 2 adalimumab levels below 4.28 and 3.37 μg/mL were predictive of primary nonresponse (area under the curve (AUC) = 0.89, 0.88; P = 0.0003, P = 0.034, respectively). Week 4 AAA signal‐to‐noise levels were significantly higher among primary nonresponders, and the cutoff for primary nonresponse prediction was above 5.31 (AUC = 0.81; P = 0.004). Adalimumab trough levels in a range of 8–12 μg/mL are optimum to reach major improvement, and lower adalimumab with higher AAA levels at the early stage (week 4) predict primary nonresponse by supporting proactive monitoring to optimize adalimumab therapy.
Collapse
Affiliation(s)
- Xiaoliang Ding
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Ruifang Zhu
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jian Wu
- Department of Rheumatology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ling Xue
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Meihua Gu
- Department of Rheumatology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liyan Miao
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
39
|
Lamers-Karnebeek FBG, Jacobs JWG, Radstake TRDJ, van Riel PLCM, Jansen TL. Adalimumab drug and antidrug antibody levels do not predict flare risk after stopping adalimumab in RA patients with low disease activity. Rheumatology (Oxford) 2020; 58:427-431. [PMID: 30383251 DOI: 10.1093/rheumatology/key292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/17/2018] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To establish whether serum adalimumab (ADA) trough level (ADA-TL) and antidrug antibody (ADA-ab) level predict flare after stopping ADA in established RA patients with long-standing low disease activity. METHODS From the clinical trial Potential Optimalisation and Effectiveness of TNF-blockers, 210 RA patients stopping ADA, who had been using ADA (40 mg/2 weeks) for >1 year with conventional synthetic DMARDs and who had low disease activity (DAS28 < 3.2, or the rheumatologist's assessment of low disease activity with CRP < 10 mg/l) for at least 6 months prior to stopping, were followed for 1 year. The ADA-TL was measured (by ELISA) 12-17 days after the last ADA injection; if it was low, ADA-abs were measured (by an antigen-binding test). Association between time-to-flare and ADA-TL was evaluated by area under the receiver operating characteristic curve and Cox regression. RESULTS A total of 106 (51%) patients flared within 1 year after stopping ADA. The area under the receiver operating characteristic curve for flare and ADA-TL was 0.50 (95% CI 0.42-0.58), P = 0.92. The hazard ratio for flare for ADA-TL ⩾ 5 μg/ml (adequate level) vs <5 μg/ml was 0.93 (95% CI: 0.63-1.36) (not significant). Of the 4 patients with high ADA-ab levels, 2 patients (50%) experienced a flare. CONCLUSION Flare risk within the year following stopping ADA is not predicted by the ADA-TL or ADA-abs assessed at the moment of stopping. TRIAL REGISTRATION Netherlands Trial Register, http://www.trialregister.nl, NTR3112.
Collapse
Affiliation(s)
| | - Johannes W G Jacobs
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Timothy R D J Radstake
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Piet L C M van Riel
- Radboud Institute for Health Sciences, IQ Healthcare, Radboud University Center, Nijmegen, The Netherlands
| | - Tim L Jansen
- Department of Rheumatology, Viecuri Medical Center, Venlo, The Netherlands
| |
Collapse
|
40
|
Ducourau E, Rispens T, Samain M, Dernis E, Le Guilchard F, Andras L, Perdriger A, Lespessailles E, Martin A, Cormier G, Armingeat T, Devauchelle-Pensec V, Gervais E, Le Goff B, de Vries A, Piver E, Paintaud G, Desvignes C, Ternant D, Watier H, Goupille P, Mulleman D. Methotrexate effect on immunogenicity and long-term maintenance of adalimumab in axial spondyloarthritis: a multicentric randomised trial. RMD Open 2020; 6:rmdopen-2019-001047. [PMID: 31958280 PMCID: PMC7046954 DOI: 10.1136/rmdopen-2019-001047] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 12/17/2022] Open
Abstract
Objectives Anti-drug antibodies (ADA) are responsible for decreased adalimumab efficacy in axial spondyloarthritis (SpA). We aimed to evaluate the ability of methotrexate (MTX) to decrease adalimumab immunisation. Methods A total of 110 patients eligible to receive adalimumab 40 mg subcutaneously (s.c.) every other week were randomised (1:1 ratio) to receive, 2 weeks before adalimumab (W-2) and weekly, MTX 10 mg s.c. (MTX+) or not (MTX−). ADA detection and adalimumab serum concentration were assessed at weeks 4 (W4), 8 (W8), 12 (W12) and 26 (W26) after starting adalimumab (W0). The primary outcome was the proportion of patients with ADA at W26. Four years after the study completion, we retrospectively analysed adalimumab maintenance in relation with MTX co-treatment duration. Results We analysed data for 107 patients (MTX+; n=52; MTX-; n=55). ADA were detected at W26 in 39/107 (36.4%) patients: 13/52 (25%) in the MTX+ group and 26/55 (47.3%) in the MTX− group (p=0.03). Adalimumab concentration was significantly higher in the MTX+ than MTX− group at W4, W8, W12 and W26. The two groups did not differ in adverse events or efficacy. In the follow-up study, MTX co-treatment >W26 versus no MTX or ≤W26 was significantly associated with adalimumab long-term maintenance (p=0.04). Conclusion MTX reduces the immunogenicity and ameliorate the pharmacokinetics of adalimumab in axial SpA. A prolonged co-treatment of MTX>W26 seems to increase adalimumab long-term maintenance.
Collapse
Affiliation(s)
- Emilie Ducourau
- Department of Rheumatology, University of Tours, EA 7501 GICC, CHRU de Tours, Tours, France.,Department of Rheumatology, CHR d'Orléans, Orléans, France
| | - Theo Rispens
- Landsteiner Laboratory, Sanquin Research, Amsterdam, Netherlands
| | - Marine Samain
- Department of Rheumatology, University of Tours, EA 7501 GICC, CHRU de Tours, Tours, France
| | | | | | - Lucia Andras
- Department of Rheumatology, CH de Blois, Blois, France
| | | | | | - Antoine Martin
- Department of Rheumatology, CH de Saint-Brieuc, Saint-Brieuc, France
| | - Grégoire Cormier
- Department of Rheumatology, CHD Vendée, La Roche-sur-Yon, France
| | - Thomas Armingeat
- Department of Rheumatology, CH de Saint-Nazaire, Saint-Nazaire, France
| | | | | | - Benoit Le Goff
- Department of Rheumatology, CHRU de Nantes, Nantes, France
| | - Annick de Vries
- Biologicals Lab, Sanquin Diagnostic Services, Amsterdam, Netherlands
| | - Eric Piver
- Department of Biochemistry, University of Tours, Inserm U 1259, CHRU de Tours, Tours, France
| | - Gilles Paintaud
- Department of Pharmacology-Toxicology, University of Tours, EA GICC, CHRU de Tours, Tours, France
| | - Céline Desvignes
- Department of Pharmacology-Toxicology, University of Tours, EA GICC, CHRU de Tours, Tours, France
| | - David Ternant
- Department of Pharmacology-Toxicology, University of Tours, EA GICC, CHRU de Tours, Tours, France
| | - Hervé Watier
- Department of Immunology, University of Tours, EA 7501 GICC, CHRU de Tours, Tours, France
| | - Philippe Goupille
- Department of Rheumatology, University of Tours, EA 7501 GICC, CHRU de Tours, Tours, France.,Inserm CIC1415, Tours, France
| | - Denis Mulleman
- Department of Rheumatology, University of Tours, EA 7501 GICC, CHRU de Tours, Tours, France
| |
Collapse
|
41
|
Abstract
The number of therapeutic antibodies available in rheumatology increases every year, mainly indicated in chronic inflammatory rheumatisms and other immune-mediated inflammatory diseases. The choice between all these monoclonal antibodies depends on their specificities, patients and diseases characteristics. Until now, there is no reliable biomarker, predictive of response for specialized medicine purpose. Today, therapeutic drug monitoring and anti-drug antibodies testing can help to better adapt the dose of the drug and to help in the decision to switch to another biological drug according to the activity of the inflammatory disease.
Collapse
Affiliation(s)
- Jacques Morel
- Département de rhumatologie, Université et CHU de Montpellier ; UMR 5535, Institut de génétique moléculaire de Montpellier (IGMM), Montpellier, France
| | - Denis Mulleman
- Université de Tours, EA 7501, Groupe insuffisance cardiaque et cardiomyopathie (GICC), CHRU de Tours, Service de rhumatologie, Tours, France
| |
Collapse
|
42
|
Sanmarti R, Veale DJ, Martin‐Mola E, Escudero‐Contreras A, González C, Ercole L, Alonso R, Fonseca JE, Alcañiz C, Álvaro‐Gracia JM, Balsa A, Pablos JL, Miguel CD, Rodríguez JM, Alves J, Aurrecoechea E, Calvo J, Belzunegui J, Blanco F, Caliz R, Calvo J, Ivorra JR, Canhão H, Santos H, Chamizo E, Pino J, Delgado C, Díaz C, Nebro AF, Fraser A, Gomez A, Hernández B, Navarro F, Povedano J, Mas AJ, Kane D, Whelan B, Marras C, Moreno J, Venegas JP, Pombo M, Riera E, Rosas A, Ryan J, Santos J, Santos M, Tornero J, Tovar JV, Ucar E, Vasconcelos C, Veiga R, Vela P. Reducing or Maintaining the Dose of Subcutaneous Tocilizumab in Patients With Rheumatoid Arthritis in Clinical Remission: A Randomized, Open‐Label Trial. Arthritis Rheumatol 2019; 71:1616-1625. [DOI: 10.1002/art.40905] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/02/2019] [Indexed: 12/20/2022]
Affiliation(s)
| | - Douglas J. Veale
- St. Vincent's University Hospital and University College Dublin Belfield Dublin Ireland
| | | | - Alejandro Escudero‐Contreras
- Rheumatology Service Reina Sofia Hospital Maimonides Institute for Research in Biomedicine of Cordoba University of Cordoba Cordoba Spain
| | | | | | | | - João E. Fonseca
- Universidade de Lisboa and Hospital de Santa Maria CHLN Lisbon Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Stamp LK, Chan SJ, Marra C, Helme C, Treharne GJ. Tapering biologic therapy for people with rheumatoid arthritis in remission: A review of patient perspectives and associated clinical evidence. Musculoskeletal Care 2019; 17:161-169. [PMID: 31148375 DOI: 10.1002/msc.1404] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/10/2019] [Accepted: 04/13/2019] [Indexed: 06/09/2023]
Abstract
OBJECTIVES Biologic therapies have increased the control of disease activity in rheumatoid arthritis (RA). Questions remain about tapering biologics when remission is achieved in RA. The patient perspective has to be incorporated in pragmatic applications of tapering but is rarely accounted for in clinical studies of tapering. The aim of the present review was to summarize the evidence about RA patient perspectives on biologic tapering. METHODS We provided a narrative summary of the currently small body of research on patient perspectives retrieved through systematic searches with an emphasis on seeking qualitative research. In addition, we provided an update on relevant clinical research and financial considerations that frame the findings on patient perspectives. RESULTS Financial considerations around commencing/continuing on biologic therapies in RA vary internationally and have implications for patient perspectives. Recent clinical studies indicate that the benefit of tapering biologic therapy when in remission are predicted by drug concentration and aspects of disease activity, severity and duration. Three major concerns have been identified from studies of patient perspectives on biologic tapering: (a) disease relapse; (b) access to treatment in the case of disease flare when tapering; and (c) local motivation for dose reduction (i.e., driven by funding or health benefit). CONCLUSIONS More research is needed on tapering biologics, and should include studies of patient perspectives as well as health economic evaluations. Patient decision aids are a potential way of applying clinical and patient-focused evidence to help all parties come to a decision, but require developmental research and pragmatic evaluation.
Collapse
Affiliation(s)
- Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, Aotearoa/New Zealand
| | - Suz Jack Chan
- School of Pharmacy, University of Otago, Dunedin, Aotearoa/New Zealand
| | - Carlo Marra
- School of Pharmacy, University of Otago, Dunedin, Aotearoa/New Zealand
| | - Caitlin Helme
- Department of Psychology, University of Otago, Dunedin, Aotearoa/New Zealand
| | - Gareth J Treharne
- Department of Psychology, University of Otago, Dunedin, Aotearoa/New Zealand
| |
Collapse
|
44
|
Dervieux T, Kremer JM, Weinblatt ME. Differing contribution of methotrexate polyglutamates to adalimumab blood levels as compared with etanercept. Ann Rheum Dis 2019; 78:1285-1286. [PMID: 30885995 PMCID: PMC6788881 DOI: 10.1136/annrheumdis-2018-214860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 02/11/2019] [Accepted: 03/04/2019] [Indexed: 12/27/2022]
Affiliation(s)
| | | | - Michael E Weinblatt
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Papamichael K, Vogelzang EH, Lambert J, Wolbink G, Cheifetz AS. Therapeutic drug monitoring with biologic agents in immune mediated inflammatory diseases. Expert Rev Clin Immunol 2019; 15:837-848. [PMID: 31180729 DOI: 10.1080/1744666x.2019.1630273] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/07/2019] [Indexed: 12/22/2022]
Abstract
Introduction: Biologic therapy has revolutionized the treatment of immune mediated inflammatory diseases (IMID), such as inflammatory bowel disease (IBD), rheumatoid and psoriatic arthritis, ankylosing spondylitis and psoriasis. Nevertheless, some patients exhibit primary nonresponse (PNR) or secondary loss of response (SLR) to biologics. Areas covered: This collaborative review provides data on the role of therapeutic drug monitoring (TDM) in IMID for optimizing biologic therapy including infliximab, adalimumab, certolizumab pegol etanercept and golimumab vedolizumab, secukinumab and ustekinumab. Expert opinion: Most exposure-response relationship studies show a positive correlation between biologic drug concentrations and favorable therapeutic outcomes in IMID with higher drug concentrations typically associated with more objective outcomes. Clinically, reactive TDM rationalizes the management of PNR and SLR to anti-tumor necrosis factor therapy and is emerging as the new standard of care in IBD as it is also more cost-effective than empiric dose escalation. Preliminary data suggest that proactive TDM with the goal to achieve a threshold drug concentration is associated with better therapeutic outcomes when compared to empiric drug optimization and/or reactive TDM of infliximab and adalimumab in IBD. However, more data from well-designed prospective studies are needed to prove the benefit of TDM-based algorithms in real life clinical practice in IMID.
Collapse
Affiliation(s)
- Konstantinos Papamichael
- a Center for Inflammatory Bowel Diseases, Division of Gastroenterology, Beth-Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA
| | - Erik H Vogelzang
- b Rheumatology and Immunology Center, Research by AMC, READE and VUMC , Amsterdam , the Netherlands
| | - Jo Lambert
- c Department of Dermatology, Ghent University , Ghent , Belgium
| | - Gertjan Wolbink
- b Rheumatology and Immunology Center, Research by AMC, READE and VUMC , Amsterdam , the Netherlands
- d Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Center , Amsterdam , the Netherlands
| | - Adam S Cheifetz
- a Center for Inflammatory Bowel Diseases, Division of Gastroenterology, Beth-Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
46
|
Jani M, Chinoy H, Barton A. Association of Pharmacological Biomarkers with Treatment Response and Longterm Disability in Patients with Psoriatic Arthritis: Results from OUTPASS. J Rheumatol 2019; 47:1204-1208. [DOI: 10.3899/jrheum.190253] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2019] [Indexed: 11/22/2022]
Abstract
Objective.To identify (1) whether tumor necrosis factor inhibitor (TNFi) drug levels/anti-drug antibodies (ADAb) are associated with treatment response and disability in patients with psoriatic arthritis (PsA); and (2) the factors associated with TNFi drug levels.Methods.Patients were recruited from a national multicenter prospective cohort with longitudinal serum samples and 28-joint count Disease Activity Scores (DAS28)/Health Assessment Questionnaire (HAQ) measurement over 12 months.Results.Adalimumab (ADA) drug levels were significantly associated with ΔDAS28 (β 0.055, 95% CI 0.011–0.099; p = 0.014) and inversely with HAQ over 12 months (β −0.022, 95% CI −0.043 to −0.00063). Factors significantly associated with ADA drug levels were ADAb levels and body mass index.Conclusion.Drug level testing in ADA-initiated PsA patients may be useful in determining treatment response/disability over 12 months.
Collapse
|
47
|
Soh MC, Moretto M. The use of biologics for autoimmune rheumatic diseases in fertility and pregnancy. Obstet Med 2019; 13:5-13. [PMID: 32284726 DOI: 10.1177/1753495x19841799] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 03/10/2019] [Indexed: 12/18/2022] Open
Abstract
In an age where autoimmune rheumatic diseases are successfully managed with biologics, their discontinuation in pregnancy is inadvisable without careful forethought; maternal disease activity is associated with adverse pregnancy outcomes, which has long-term implications for both mother and offspring. We aim to provide clinicians with the necessary tools to facilitate decision-making - when a biologic should be used, when it can be discontinued in pregnancy if appropriate. The pathophysiology of these biologic molecules and their effect on fertility, pregnancy and parturition are discussed. A summary of the 2016 international guidelines (European League Against Rheumatism and British Society in Rheumatology) on biologics in pregnancy has been tabulated; more recent publications are discussed in depth. Data on transplacental-transfer ratios and breastmilk excretion rates are also included. Biologic effects on organogenesis, their implications for the exposed infant in terms of infection risks and vaccination requirements are included, and future directions for research proposed.
Collapse
Affiliation(s)
- May Ching Soh
- Department of Rheumatology, Tauranga Hospital, Bay of Plenty District Health Board, Tauranga, New Zealand.,Department of Obstetrics and Gynaecology, Elizabeth Rothwell Building, Waikato Hospital, Waikato District Health Board, Hamilton, New Zealand.,Women's Health Academic Centre, King's College London, London, UK
| | - Marcelo Moretto
- Department of Gynaecology, Hospital São Lucas da Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Clínica Generar-Human Reproduction, Porto Alegre, Brazil
| |
Collapse
|
48
|
Jani M, Barton A, Hyrich K. Prediction of infection risk in rheumatoid arthritis patients treated with biologics: are we any closer to risk stratification? Curr Opin Rheumatol 2019; 31:285-292. [PMID: 30789850 PMCID: PMC6443047 DOI: 10.1097/bor.0000000000000598] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW There are currently several available biologics for rheumatoid arthritis (RA) with similar efficacy in most trials. A major consideration therefore in choosing a biologic, continues to be safety concerns such as infection. Considerable advances have been made in the understanding of biologic safety on a population level; however, how close are we to stratifying risk for individual patients? This review discusses evidence published in the last year, with reference to key previous literature. RECENT FINDINGS Comparative safety of biologics has been studied in observational cohorts, with a possible increased risk of serious infection in tocilizumab-treated patients compared with etanercept. Rheumatoid arthritis patients on biologics are often on concomitant medications such as steroids and opioids, and the advances in relation to infection are summarized. Pharmacological biomarkers and optimizing existing risk prediction scores may allow better future risk stratification. SUMMARY Improved quantification of personalized benefit:harms would allow better-informed decisions, reduction of infection-associated morbidity as well as direct/indirect costs associated with biologics. Although advances have been made to better understand and predict risk, future studies are likely to require a range of novel data sources and methodologies for the goal of precision medicine to be truly realized.
Collapse
Affiliation(s)
- Meghna Jani
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, The University of Manchester
- Rheumatology Department, Salford Royal NHS Foundation Trust, Salford, UK
| | - Anne Barton
- Arthritis Research UK Centre for Genetics and Genomics, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester
| | - Kimme Hyrich
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, The University of Manchester
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester
| |
Collapse
|
49
|
L' Ami MJ, Ruwaard J, Krieckaert C, Nurmohamed MT, van Vollenhoven RF, Rispens T, Wolbink GJ. Serum drug concentrations to optimize switching from adalimumab to etanercept in rheumatoid arthritis. Scand J Rheumatol 2019; 48:266-270. [PMID: 31012365 DOI: 10.1080/03009742.2019.1577915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objectives: Inadequate response to adalimumab can be caused by insufficient blockade of the target tumour necrosis factor (TNF) at low serum concentrations. In such cases, patients may respond to another TNF inhibitor. We investigated whether the serum adalimumab concentration is related to the efficacy of a second TNF inhibitor, etanercept, in rheumatoid arthritis (RA). Methods: Patients with RA starting etanercept treatment were prospectively observed in the Reade Rheumatology Registry. In patients previously on adalimumab, serum concentrations were determined before treatment discontinuation. According to this concentration, three subgroups were formed: < 0.5 μg/mL, 0.5-5.0 μg/mL, and ≥ 5.0 μg/mL. The European League Against Rheumatism (EULAR) good/moderate response rate after 52 weeks of etanercept was compared between the switcher subgroups and biologic-naive patients. Results: In total, 449 consecutive patients were included, of whom 69 switched from adalimumab (15%) and 380 were biologic naive (85%). EULAR good or moderate response was achieved by 74% of the biologic-naive patients and by 72%, 50%, and 52% of switchers with adalimumab concentration < 0.5 μg/mL, 0.5-5.0 μg/mL, and ≥ 5.0 μg/mL, respectively (p = 0.15). Patients with an adalimumab concentration ≥ 0.5 μg/mL were significantly less likely to achieve EULAR good/moderate response on etanercept compared to biologic-naive patients, whereas patients with a concentration < 0.5 μg/mL did not significantly differ from patients starting etanercept without prior biologic treatment. Conclusion: RA patients with an inadequate response to adalimumab, in the presence of sufficient drug concentrations, benefit less from switching to another TNF inhibitor, etanercept.
Collapse
Affiliation(s)
- M J L' Ami
- a Department of Rheumatology , Amsterdam Rheumatology and Immunology Center , Reade , Amsterdam , The Netherlands
| | - J Ruwaard
- a Department of Rheumatology , Amsterdam Rheumatology and Immunology Center , Reade , Amsterdam , The Netherlands
| | - Clm Krieckaert
- a Department of Rheumatology , Amsterdam Rheumatology and Immunology Center , Reade , Amsterdam , The Netherlands
| | - M T Nurmohamed
- a Department of Rheumatology , Amsterdam Rheumatology and Immunology Center , Reade , Amsterdam , The Netherlands.,b Department of Rheumatology , Amsterdam Rheumatology and Immunology Center, UMC/VU University Medical Center , Amsterdam , The Netherlands
| | - R F van Vollenhoven
- a Department of Rheumatology , Amsterdam Rheumatology and Immunology Center , Reade , Amsterdam , The Netherlands.,b Department of Rheumatology , Amsterdam Rheumatology and Immunology Center, UMC/VU University Medical Center , Amsterdam , The Netherlands.,c Department of Rheumatology , Amsterdam Rheumatology and Immunology Center, UMC/Academic Medical Center , Amsterdam , The Netherlands
| | - T Rispens
- d Department of Immunopathology , Sanquin Research and Landsteiner Laboratory Academic Medical Center , Amsterdam , The Netherlands
| | - G J Wolbink
- a Department of Rheumatology , Amsterdam Rheumatology and Immunology Center , Reade , Amsterdam , The Netherlands.,d Department of Immunopathology , Sanquin Research and Landsteiner Laboratory Academic Medical Center , Amsterdam , The Netherlands
| |
Collapse
|
50
|
Renton WD, Ramanan AV. Biological therapeutic drug monitoring: a step towards precision medicine? Arch Dis Child 2019; 104:212-213. [PMID: 30262510 DOI: 10.1136/archdischild-2018-315819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/14/2018] [Accepted: 08/31/2018] [Indexed: 11/03/2022]
Affiliation(s)
- William D Renton
- Bristol Royal Hospital for Children, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Athimalaipet V Ramanan
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|