1
|
Sarsarshahi S, Bhattacharya S, Zacharias ZR, Kamel ES, Houtman JCD, Nejadnik R. Highly variable aggregation and glycosylation profiles and their roles in immunogenicity to protein-based therapeutics. J Pharm Sci 2025; 114:103771. [PMID: 40139530 DOI: 10.1016/j.xphs.2025.103771] [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: 02/11/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Production of antibodies against protein-based therapeutics (e.g., monoclonal antibodies (mAbs)) by a recipient's immune system can vary from benign symptoms to chronic neutralization of the compound, and in rare cases, a lethal cytokine storm. One critical factor that can induce or contribute to an anti-drug antibody (ADA) response is believed to be the presence of aggregated proteins in protein-based therapeutics. There is a high level of variability in the aggregation of different proteins, which adds to the complexity in understanding the immune response to these drugs. Furthermore, the level of glycosylation of proteins, which increases drug stability, functionality, and serum half-life, is highly variable and may influence their immunogenicity. Considering the abundance of literature on the effect of aggregation and glycosylation on the immunogenicity of protein-based therapeutics, this review aims to summarize the current knowledge and clarify the immunogenic effects of different protein-based therapeutics such as mAbs. This review focuses on the properties of aggregated proteins and elucidates their relationship with immunogenicity. The contribution of different immune cell subsets and the mechanisms in aggregation-induced immunogenicity are also reviewed. Finally, the potential effects of each glycan, such as sialic acid, mannose, and fucose, on protein-based therapeutics' immunogenicity and stability is discussed.
Collapse
Affiliation(s)
- Sina Sarsarshahi
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Sanghati Bhattacharya
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Zeb R Zacharias
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States
| | - Eman S Kamel
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Jon C D Houtman
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Reza Nejadnik
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
2
|
Di Santo A, Accinno M, Errante F, Capone M, Vultaggio A, Simoncini E, Zipoli G, Cosmi L, Annunziato F, Rovero P, Real Fernandez F. Quantitative evaluation of adalimumab and anti-adalimumab antibodies in sera using a surface plasmon resonance biosensor. Clin Biochem 2024; 133-134:110838. [PMID: 39489392 DOI: 10.1016/j.clinbiochem.2024.110838] [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: 07/13/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVES Monitoring of therapeutic antibody adalimumab (ADL) and of anti-adalimumab antibodies (AAA) in autoimmune diseases patients' sera has achieved increased attention since several studies showed a correlation between AAA levels and treatment failure. We evaluated a new surface plasmon resonance (SPR)-based method that, with slight changes in the analysis condition and in the ligand immobilized on the chip surface, allows to monitor both AAA and ADL. This new label-free method does not require sample pretreatments, and it is fully automated, only requiring the preparation of the chip, which can be used for multiple analysis, and the preparation of the sample sets. DESIGN & METHODS Sera from ADL-treated patients (n = 47) and controls (n = 13) were included in this study. Quantitative analysis of AAA and ADL were performed separately using a new SPR-biosensor, and a commercially available ELISA kit. Agreement was defined by overall, positive, and negative agreement. Wilson Score was used to calculate confidence intervals (CI) on binomial probability and Spearman's rho and Bland-Altman test were used to assess correlations. RESULTS ELISA and SPR-based assay were able to identify circulating AAA in ADL-treated patients, with the percentage of positivity varying among the methods, with an overall agreement of 79%. AAA were detected in 18 (38 %) out of the 47 treated patients by the ELISA whereas SPR-based assay detected 10 (21 %) out of 47 samples. CONCLUSIONS Real-time label free SPR-based protocol for both AAA and ADL quantification has been set-up. Although quantitative differences were observed when compared with ELISA, the agreement among methodologies was high, particularly for ADL quantification within the therapeutic window of the drug.
Collapse
Affiliation(s)
- Andrea Di Santo
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of NeuroFarBa, University of Florence, Sesto Fiorentino, Italy
| | - Matteo Accinno
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Fosca Errante
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of NeuroFarBa, University of Florence, Sesto Fiorentino, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Department of Laboratory Medicine, Azienda Usl Toscana Centro, Florence, Italy
| | - Alessandra Vultaggio
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | - Eleonora Simoncini
- Clinical Trials Task Force - Ethics and Care Unit, Azienda Usl Toscana Centro, Florence, Italy
| | - Giuditta Zipoli
- Clinical Trials Task Force - Ethics and Care Unit, Azienda Usl Toscana Centro, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Paolo Rovero
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of NeuroFarBa, University of Florence, Sesto Fiorentino, Italy.
| | - Feliciana Real Fernandez
- Istituto di Chimica dei Composti Organometallici (ICCOM), Consiglio Nazionale delle Ricerche (CNR), Sesto Fiorentino (FI) I-50019, Italy.
| |
Collapse
|
3
|
Keating PE, Hock BD, Chin PKL, O'Donnell JL, Barclay ML. Evaluation of the Homogenous Mobility Shift Assay for Infliximab and Adalimumab Anti-drug Antibody Detection in the Clinical Laboratory. Ther Drug Monit 2024; 46:619-626. [PMID: 38648648 DOI: 10.1097/ftd.0000000000001200] [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: 08/02/2023] [Accepted: 02/08/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Detecting antidrug antibodies (ADAs) against infliximab or adalimumab is useful for therapeutic drug monitoring. Various ADA detection methods exist, and antibody titer is an output in some algorithms. Homogenous mobility shift assay (HMSA) measures relative ADA concentration and determines drug-ADA complex size in vitro. However, the relevance of complex size determination in drug monitoring remains unclear. Hence, the association between complex size, ADA concentration, and sample detectable neutralizing activity was evaluated. METHODS Sera from infliximab-treated and adalimumab-treated patients who tested positive for ADA in the National Screening Service were analyzed using 3 ADA assays. HMSA determined the relative ADA concentrations and complex sizes, competitive ligand-binding assay evaluated the sample neutralizing capacity, and enzyme-linked immunosorbent assay detected immunoglobulin (Ig)G4 ADA. RESULTS Most ADA-positive samples (>80%) formed drug-ADA dimer complexes, whereas 17% had dimer and multimer complexes, and 3% had multimeric complexes. Multimer presence had 100% positive predictive value for detectable neutralizing activity. ADA concentration and detectable neutralizing activity were moderately correlated (r = 0.65) in adalimumab-treated patients and strongly correlated (r = 0.81) in infliximab-treated patients. In adalimumab-treated patients, multimer presence was a stronger predictor of neutralizing activity than ADA concentration was, but not in infliximab-treated patients. However, in infliximab-treated patient samples, multimer presence revealed a distinct subset with high ADA concentrations, neutralizing activity, and IgG4 ADA. CONCLUSIONS Multimers detected using HMSA had a strong positive predictive value for competitive ligand-binding assay detectable neutralizing activity. Multimeric IgG4-containing ADA-drug complexes revealed a distinct subset of infliximab-treated patient samples, whose clinical relevance merits further investigation.
Collapse
Affiliation(s)
| | - Barry D Hock
- Department of Hematology, University of Otago, Christchurch, New Zealand
| | - Paul K L Chin
- Department of Medicine, University of Otago, Christchurch, New Zealand ; and
- Department of Clinical Pharmacology, Christchurch Hospital, Christchurch, New Zealand
| | | | - Murray Lindsay Barclay
- Department of Medicine, University of Otago, Christchurch, New Zealand ; and
- Department of Clinical Pharmacology, Christchurch Hospital, Christchurch, New Zealand
| |
Collapse
|
4
|
Sun M, Ju J, Xu H, Luo M, Li Z, Wang Y. Antibiotics influence the risk of anti-drug antibody formation during anti-TNF therapy in Chinese inflammatory bowel disease patients. Front Pharmacol 2024; 15:1360835. [PMID: 38655181 PMCID: PMC11035825 DOI: 10.3389/fphar.2024.1360835] [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: 12/24/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024] Open
Abstract
Aims: The formation of anti-drug antibodies (ADAs) during anti-tumor necrosis factor (anti-TNF) therapy is reported to lead to reducing serum drug levels, which may bring about a loss of response to treatment. Previous research has suggested an association between specific antibiotic classes and ADA formation during anti-TNF therapy. However, there are few studies specifically examining this association in Chinese inflammatory bowel disease (IBD) patients. Therefore, our study aimed to evaluate the possible effect of antibiotic use on ADA formation to anti-TNF therapy in Chinese patients with IBD. Methods: A total of 166 patients with IBD, including 149 with Crohn's disease (CD) and 17 with ulcerative colitis (UC), were included in this retrospective analysis. These patients were initially treated with anti-TNF therapy (infliximab or adalimumab) after January 2018 and reviewed with available ADA levels before October 2023. After univariable analysis of all the variables, a multivariate Cox proportional hazards model was used to assess the association between antibiotic use and ADA development. Results: Among 166 IBD patients treated with infliximab (108/166, 65.1%) or adalimumab (58/166, 34.9%), 31 patients (18.7%) were measured as positive ADA levels. Cox proportional hazard model demonstrated an increased risk of ADA formation in IBD patients who used β-lactam-β-lactamase inhibitor combinations (BL-BLIs) (HR = 5.143, 95%CI 1.136-23.270, p = 0.033), or nitroimidazoles (HR = 4.635, 95%CI 1.641-13.089, p = 0.004) during 12 months before the ADA test. On the contrary, a reduced risk was noted in patients treated with fluoroquinolones (HR = 0.258, 95% CI 0.072-0.924, p = 0.037). Moreover, the median serum infliximab or adalimumab concentration in patients with positive ADA levels was significantly lower than that in patients with negative ADA levels (infliximab: 0.30 vs. 1.85 μg/mL, p < 0.0001; adalimumab: 0.45 vs. 7.55 μg/mL, p = 0.0121). Conclusion: ADA development is associated with various antibiotic classes. BL-BLIs and nitroimidazoles might increase the risk of ADA formation during anti-TNF therapy in Chinese IBD patients, while the treatment with fluoroquinolones could probably reduce such risk. There were certain limitations in the retrospective analysis of the study, therefore, the results are just for reference, and other studies are needed to further confirm our findings.
Collapse
Affiliation(s)
| | | | | | | | | | - Yufang Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Barrau M, Duprat M, Veyrard P, Tournier Q, Williet N, Marc Phelip J, Waeckel L, Cheifetz AS, Papamichael K, Roblin X, Paul S. A Systematic Review on the Interest of Drug-tolerant Assay in the Monitoring of Inflammatory Bowel Disease. J Crohns Colitis 2023; 17:633-643. [PMID: 36301958 DOI: 10.1093/ecco-jcc/jjac164] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Indexed: 02/08/2023]
Abstract
Many patients with inflammatory bowel disease [IBD] are treated with anti-tumour necrosis factor [TNF] therapies, of which infliximab [IFX] is most commonly used. Loss of response [LOR] to anti-TNF therapy due to immunogenic failure accounts for 20% of subsequent medical intervention and is defined, using a drug-sensitive assay, as low or undetectable concentration of drug with high titres of anti-drug antibodies [ADAb]. We performed a systematic review to investigate the use of a drug-tolerant assay during both induction and maintenance, to monitor patients treated with anti-TNFs. After the search on PubMed, 90 publications were reviewed. Most ADAb detection methods are drug-sensitive, cannot detect ADAb in the presence of drug, and therefore cannot be used close to drug administration when the drug concentration is too high. To overcome this major limitation, several drug-tolerant techniques have been developed and will be discussed in this review. Using drug-tolerant assays, ADAb against IFX or adalimumab [ADM] can be detected during induction and predict primary non-response or LOR. Drug-sensitive assays do not allow detection of ADAb during the induction phase when IFX or ADM concentration is typically high.
Collapse
Affiliation(s)
- Mathilde Barrau
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Manon Duprat
- Department of Immunology, Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Saint-Etienne, France
| | - Pauline Veyrard
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Quentin Tournier
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Nicolas Williet
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Jean Marc Phelip
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Louis Waeckel
- Department of Immunology, Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Saint-Etienne, France
| | - Adam S Cheifetz
- Center for Inflammatory Bowel Disease, Beth Israel Deaconess Medical Center , Harvard Medical School, Boston, MA, USA
| | - Konstantinos Papamichael
- Center for Inflammatory Bowel Disease, Beth Israel Deaconess Medical Center , Harvard Medical School, Boston, MA, USA
| | - Xavier Roblin
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Stephane Paul
- Department of Immunology, Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Saint-Etienne, France
| |
Collapse
|
6
|
Terabe K, Takahashi N, Asai S, Hirano Y, Kanayama Y, Yabe Y, Oguchi T, Fujibayashi T, Ishikawa H, Hanabayashi M, Hattori Y, Suzuki M, Kishimoto K, Ohashi Y, Imaizumi T, Imagama S, Kojima T. Effectiveness of tacrolimus concomitant with biological disease-modifying antirheumatic drugs in patients with rheumatoid arthritis. Mod Rheumatol 2023; 33:292-301. [PMID: 35289372 DOI: 10.1093/mr/roac025] [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: 11/09/2021] [Revised: 01/22/2022] [Accepted: 02/09/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVES The study aimed to investigate the effectiveness and tolerance of biological disease-modifying antirheumatic drugs (bDMARDs) therapy administered concomitantly with tacrolimus (TAC) treatment in patients with rheumatoid arthritis. METHODS 2792 patients who underwent therapy with five bDMARDs (etanercept: ETN, adalimumab, golimumab, tocilizumab, and abatacept: ABT) were enrolled. Among the study subjects, 1582 were concomitant methotrexate (MTX group), 147 were concomitant TAC (TAC group), and 1063 were non-concomitant MTX and TAC (non-MTX/TAC group). The primary outcome was the incident rate of discontinuation of bDMARDs by adverse events (AEs) or loss of efficacy. RESULTS Concerning the analysis for each reasons of discontinuation, including AEs and loss of efficacy, the hazards ratio (HR) was significantly lower in the TAC group than in non-MTX/TAC groups (AEs: HR = 0.39, 95% confidence interval, 0.23-0.68, loss of efficacy: HR = 0.49, 95% confidence interval, 0.30-0.78). The loss of efficacy with the use of ETN and ABT was lower in the TAC group than in non-MTX/TAC groups. Concomitant TAC did not induce elevated risk for discontinuation of AEs in all bDMARD analyses. CONCLUSIONS Concomitant TAC with ABT or ETN showed higher retention rates than bDMARDs therapy without TAC or MTX. AEs did not increase over long-term observation.
Collapse
Affiliation(s)
- Kenya Terabe
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobunori Takahashi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shuji Asai
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuji Hirano
- Department of Rheumatology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Yasuhide Kanayama
- Department of Orthopedic Surgery, Toyota Kosei Hospital, Toyota, Japan
| | - Yuichiro Yabe
- Department of Rheumatology, Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Takeshi Oguchi
- Department of Orthopedic Surgery, Anjo Kosei Hospital, Anjo, Japan
| | | | - Hisato Ishikawa
- Department of Orthopedic Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan
| | - Masahiro Hanabayashi
- Department of Orthopedic Surgery, Ichinomiya Municipal Hospital, Ichinomiya, Japan
| | - Yosuke Hattori
- Department of Orthopedic Surgery, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Mochihito Suzuki
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Kishimoto
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshifumi Ohashi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Imaizumi
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshihisa Kojima
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
7
|
Rodríguez Azor B, Martín-Masot R, Dayaldasani Khialani A, Fernández-Martín JM, Gallego Fernández C, Navas-López VM. Proactive monitoring of anti-TNF agents improves follow-up of paediatric patients with Crohn disease. An Pediatr (Barc) 2023; 98:165-174. [PMID: 36804330 DOI: 10.1016/j.anpede.2023.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/20/2022] [Indexed: 02/19/2023] Open
Abstract
INTRODUCTION AND AIMS The incidence of paediatric inflammatory bowel disease has increased in recent decades. The aim of the present study was to evaluate the role of proactive and serial monitoring of tumour necrosis factor (TNF) inhibitor levels to maintain clinical remission and mucosal healing in the follow-up of paediatric patients with Crohn disease (CD). METHODS Prospective study that included all patients diagnosed with CD and treated with adalimumab or infliximab between May 2015 and November 2020 who underwent serial and proactive monitoring of TNF inhibitor levels. RESULTS The study included 30 patients, 21 male (70%). The mean age at diagnosis was 11.3 years (SD, 2.0), the mean age at initiation of TNF inhibitors was 12.6 years (SD, 1.9) with a mean duration of follow-up of 27.1 ± 9.1 months. Clinical remission was defined as a weighted Pediatric Crohn's Disease Activity Index (wPCDAI) of less than 12.5 and mucosal healing as a Mucosal Inflammation Non-invasive Index (MINI) of less than 8. During the follow-up, patients were in clinical remission in 87.1% of the visits, presented with mild disease in 11.4% and with moderate disease in 1.5%, and mucosal healing was assumed in 83% of the visits. The rates of clinical remission and mucosal healing at 1, 2, and 3 years of follow-up were 83.3%, 95.8%, 92.8%, and 86.7%, 87.5% and 85.7%, respectively. CONCLUSIONS Proactive and serial monitoring of serum TNF inhibitor levels may make it possible for patients to maintain clinical remission and mucosal healing in the maintenance phase, with individualised optimization of the required dosage and minimization of secondary loss of response.
Collapse
Affiliation(s)
- Begoña Rodríguez Azor
- Sección de Gastroenterología y Nutrición Infantil, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Rafael Martín-Masot
- Sección de Gastroenterología y Nutrición Infantil, Hospital Regional Universitario de Málaga, Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain.
| | | | | | | | - Víctor Manuel Navas-López
- Sección de Gastroenterología y Nutrición Infantil, Hospital Regional Universitario de Málaga, Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| |
Collapse
|
8
|
La monitorización proactiva de niveles de anti-TNF mejora el seguimiento de los pacientes pediátricos con enfermedad de Crohn. An Pediatr (Barc) 2023. [DOI: 10.1016/j.anpedi.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
9
|
Curci D, Lucafò M, Parisse P, Decorti G, Bramuzzo M, Casalis L, Stocco G. Atomic Force Microscopy Application for the Measurement of Infliximab Concentration in Healthy Donors and Pediatric Patients with Inflammatory Bowel Disease. J Pers Med 2022; 12:948. [PMID: 35743733 PMCID: PMC9225523 DOI: 10.3390/jpm12060948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/26/2022] [Accepted: 06/08/2022] [Indexed: 01/14/2023] Open
Abstract
The use of infliximab has completely changed the therapeutic landscape in inflammatory bowel disease. However, despite its proven efficacy to induce and maintain clinical remission, increasing evidence suggests that treatment failure may be associated with inadequate drug blood concentrations. The introduction of biosensors based on different nanostructured materials for the rapid quantification of drugs has been proposed for therapeutic drug monitoring. This study aimed to apply atomic force microscopy (AFM)-based nanoassay for the measurement of infliximab concentration in serum samples of healthy donors and pediatric IBD patients. This assay measured the height signal variation of a nanostructured gold surface covered with a self-assembled monolayer of alkanethiols. Inside this monolayer, we embedded the DNA conjugated with a tumor necrosis factor able to recognize the drug. The system was initially fine-tuned by testing known infliximab concentrations (0, 20, 30, 40, and 50 nM) in buffer and then spiking the same concentrations of infliximab into the sera of healthy donors, followed by testing pediatric IBD patients. A good correlation between height variation and drug concentration was found in the buffer in both healthy donors and pediatric IBD patients (p-value < 0.05), demonstrating the promising use of AFM nanoassay in TDM.
Collapse
Affiliation(s)
- Debora Curci
- Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (D.C.); (M.L.); (G.D.); (M.B.); (G.S.)
| | - Marianna Lucafò
- Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (D.C.); (M.L.); (G.D.); (M.B.); (G.S.)
| | - Pietro Parisse
- Elettra Sincrotrone Trieste S.C.p.A., 34137 Trieste, Italy;
- Istituto Officina dei Materiali—CNR, 34149 Trieste, Italy
| | - Giuliana Decorti
- Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (D.C.); (M.L.); (G.D.); (M.B.); (G.S.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Matteo Bramuzzo
- Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (D.C.); (M.L.); (G.D.); (M.B.); (G.S.)
| | | | - Gabriele Stocco
- Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (D.C.); (M.L.); (G.D.); (M.B.); (G.S.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
10
|
Cui G, Florholmen J, Goll R. Could Mucosal TNF Transcript as a Biomarker Candidate Help Optimize Anti-TNF Biological Therapy in Patients With Ulcerative Colitis? Front Immunol 2022; 13:881112. [PMID: 35663996 PMCID: PMC9162116 DOI: 10.3389/fimmu.2022.881112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022] Open
Abstract
Anti-tumor necrosis factor (TNF) biological therapy has generally been accepted as a standard therapeutic option in inflammatory bowel disease (IBD) patient who are refractory to steroids or immunomodulators. However, the primary and secondary nonresponse rates to anti-TNF bioagents in patients with IBD are high. To improve the response rate, anti-TNF bioagents must be offered to the appropriate IBD patients, and the withdrawal of anti-TNF bioagents needs to be done at the right time. In this context, reliable and reproducible biomarkers can provide important supportive information for clinicians to make correct decisions based on the patient’s individual situation. In this review, we summarized the current understanding of using mucosal TNF transcript (TNF) to improve the precision of anti-TNF biological therapy strategies in patients with ulcerative colitis (UC). Analysis of published literature showed that mucosal TNF could affect the precision of the early identification of candidates who will benefit from anti-TNF therapy prior to treatment, the assessment of response and mucosal healing, and the prediction of discontinuation of anti-TNF biological therapy and relapse after drug withdrawal. Challenges and limitations of using mucosal TNF as a biomarker in applying individualized anti-TNF biological therapy in patients with UC still remain and need to be further investigated.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Faculty of Health Science, Nord University, Campus Levanger, Levanger, Norway.,Division of Gastroenterology, Department of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Jon Florholmen
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Rasmus Goll
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
11
|
Hiltunen J, Parmanne P, Sokka T, Lamberg T, Isomäki P, Kaipiainen-Seppänen O, Peltomaa R, Uutela T, Pirilä L, Taimen K, Kauppi MJ, Yli-Kerttula T, Tuompo R, Relas H, Kortelainen S, Paalanen K, Asikainen J, Ekman P, Santisteban A, Vidqvist KL, Tadesse K, Romu M, Borodina J, Elfving P, Valleala H, Leirisalo-Repo M, Rantalaiho V, Kautiainen H, Jokiranta TS, Eklund KK. Immunogenicity of subcutaneous TNF inhibitors and its clinical significance in real-life setting in patients with spondyloarthritis. Rheumatol Int 2022; 42:1015-1025. [PMID: 34357455 PMCID: PMC9124652 DOI: 10.1007/s00296-021-04955-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/17/2021] [Indexed: 01/17/2023]
Abstract
KEY MESSAGES Considerable proportion of patients with SpA have been immunized to the subcutaneous anti-TNF drug they are using. Concomitant use of MTX protects from immunization, whereas SASP does not. Patients with SpA using subcutaneous anti-TNF drugs can benefit from monitoring of the drug trough levels. Immunization to biological drugs can lead to decreased efficacy and increased risk of adverse effects. The objective of this cross-sectional study was to assess the extent and significance of immunization to subcutaneous tumor necrosis factor (TNF) inhibitors in axial spondyloarthritis (axSpA) patients in real-life setting. A serum sample was taken 1-2 days before the next drug injection. Drug trough concentrations, anti-drug antibodies (ADAb) and TNF-blocking capacity were measured in 273 patients with axSpA using subcutaneous anti-TNF drugs. The clinical activity of SpA was assessed using the Bath AS Disease Activity Index (BASDAI) and the Maastricht AS Entheses Score (MASES). ADAb were found in 11% of the 273 patients: in 21/99 (21%) of patients who used adalimumab, in 0/83 (0%) of those who used etanercept, in 2/79 (3%) of those who used golimumab and in 6/12 (50%) of those who used certolizumab pegol. Use of methotrexate reduced the risk of formation of ADAb, whereas sulfasalazine did not. Presence of ADAb resulted in decreased drug concentration and reduced TNF-blocking capacity. However, low levels of ADAb had no effect on TNF-blocking capacity and did not correlate with disease activity. The drug trough levels were below the consensus target level in 36% of the patients. High BMI correlated with low drug trough concentration. Patients with low drug trough levels had higher disease activity. The presence of anti-drug antibodies was associated with reduced drug trough levels, and the patients with low drug trough levels had higher disease activity. The drug trough levels were below target level in significant proportion of patients and, thus, measuring the drug concentration and ADAb could help to optimize the treatment in SpA patients.
Collapse
Affiliation(s)
- J. Hiltunen
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - P. Parmanne
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - T. Sokka
- Department of Rheumatology, Jyväskylä Central Hospital, Jyväskylä, Finland
- University of Eastern Finland, Kuopio, Finland
| | - T. Lamberg
- United Medix Laboratories, Helsinki, Finland
| | - P. Isomäki
- Department of Rheumatology, Tampere University Hospital, Tampere, Finland
| | | | - R. Peltomaa
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - T. Uutela
- Department of Rheumatology, Central Hospital of Lapland, Rovaniemi, Finland
| | - L. Pirilä
- Department of Rheumatology, Turku University Hospital, Turku, Finland
| | - K. Taimen
- Department of Rheumatology, Turku University Hospital, Turku, Finland
| | - M. J. Kauppi
- Department of Rheumatology, Päijät-Häme Central Hospital, Lahti, Finland
- University of Tampere, Tampere, Finland
| | - T. Yli-Kerttula
- Department of Rheumatology, Satakunta Central Hospital, Rauma, Finland
| | - R. Tuompo
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - H. Relas
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - S. Kortelainen
- Department of Rheumatology, Turku University Hospital, Turku, Finland
| | - K. Paalanen
- Department of Rheumatology, Jyväskylä Central Hospital, Jyväskylä, Finland
- University of Eastern Finland, Kuopio, Finland
| | - J. Asikainen
- Department of Rheumatology, Jyväskylä Central Hospital, Jyväskylä, Finland
- University of Eastern Finland, Kuopio, Finland
| | - P. Ekman
- Department of Rheumatology, Satakunta Central Hospital, Rauma, Finland
| | - A. Santisteban
- Department
of Rheumatology, Mikkeli Central Hospital, Mikkeli, Finland
| | - K.-L. Vidqvist
- Department of Rheumatology, Tampere University Hospital, Tampere, Finland
| | - K. Tadesse
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - M. Romu
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - J. Borodina
- Department of Rheumatology, Jyväskylä Central Hospital, Jyväskylä, Finland
- University of Eastern Finland, Kuopio, Finland
| | - P. Elfving
- Department of Rheumatology, Kuopio University Hospital, Kuopio, Finland
| | - H. Valleala
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - M. Leirisalo-Repo
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
| | - V. Rantalaiho
- Department of Rheumatology, Tampere University Hospital, Tampere, Finland
| | | | | | - K. K. Eklund
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, Haartmaninkatu 4, P. O. Box 372, 00029 HUS Helsinki, Finland
- Translational Immunology Research Program, Helsinki University and Orton Research Foundation, Orton Hospital, Helsinki, Finland
| |
Collapse
|
12
|
Ishiwatari-Ogata C, Kyuuma M, Ogata H, Yamakawa M, Iwata K, Ochi M, Hori M, Miyata N, Fujii Y. Ozoralizumab, a Humanized Anti-TNFα NANOBODY ® Compound, Exhibits Efficacy Not Only at the Onset of Arthritis in a Human TNF Transgenic Mouse but Also During Secondary Failure of Administration of an Anti-TNFα IgG. Front Immunol 2022; 13:853008. [PMID: 35273620 PMCID: PMC8902368 DOI: 10.3389/fimmu.2022.853008] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 12/19/2022] Open
Abstract
Although the introduction of tumor necrosis factor (TNF) inhibitors represented a significant advance in the treatment of rheumatoid arthritis (RA), traditional anti-TNFα antibodies are somewhat immunogenic, and their use results in the formation of anti-drug antibodies (ADAs) and loss of efficacy (secondary failure). Ozoralizumab is a trivalent, bispecific NANOBODY® compound that differs structurally from IgGs. In this study we investigated the suppressant effect of ozoralizumab and adalimumab, an anti-TNFα IgG, on arthritis and induction of ADAs in human TNF transgenic mice. Ozoralizumab markedly suppressed arthritis progression and did not induce ADAs during long-term administration. We also developed an animal model of secondary failure by repeatedly administering adalimumab and found that switching from adalimumab to ozoralizumab was followed by superior anti-arthritis efficacy in the secondary-failure animal model. Moreover, ozoralizumab did not form large immune complexes that might lead to ADA formation. The results of our studies suggest that ozoralizumab, which exhibited low immunogenicity in the animal model used and has a different antibody structure from that of IgGs, is a promising candidate for the treatment of RA patients not only at the onset of RA but also during secondary failure of anti-TNFα treatment.
Collapse
Affiliation(s)
| | - Masanao Kyuuma
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Hitoshi Ogata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Machi Yamakawa
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Katsuya Iwata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Motoki Ochi
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Miyuki Hori
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Noriyuki Miyata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Yasuyuki Fujii
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| |
Collapse
|
13
|
Suh K, Kyei I, Hage DS. Approaches for the detection and analysis of anti-drug antibodies to biopharmaceuticals: A review. J Sep Sci 2022; 45:2077-2092. [PMID: 35230731 DOI: 10.1002/jssc.202200112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/10/2022] [Accepted: 02/26/2022] [Indexed: 11/10/2022]
Abstract
Antibody-based therapeutic agents and other biopharmaceuticals are now used in the treatment of many diseases. However, when these biopharmaceuticals are administrated to patients, an immune reaction may occur that can reduce the drug's efficacy and lead to adverse side effects. The immunogenicity of biopharmaceuticals can be evaluated by detecting and measuring antibodies that have been produced against these drugs, or anti-drug antibodies (ADAs). Methods for ADA detection and analysis can be important during the selection of a therapeutic approach based on such drugs and is crucial when developing and testing new biopharmaceuticals. This review examines approaches that have been used for ADA detection, measurement, and characterization. Many of these approaches are based on immunoassays and antigen binding tests, including homogeneous mobility shift assays. Other techniques that have been used for the analysis of ADAs are capillary electrophoresis, reporter gene assays, surface plasmon resonance spectroscopy, and liquid chromatography-mass spectrometry. The general principles of each approach will be discussed, along with their recent applications with regards to ADA analysis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kyungah Suh
- Department of Chemistry, University of Nebraska-Lincoln
| | - Isaac Kyei
- Department of Chemistry, University of Nebraska-Lincoln
| | - David S Hage
- Department of Chemistry, University of Nebraska-Lincoln
| |
Collapse
|
14
|
Genaro LM, Gomes LEM, Franceschini APMDF, Ceccato HD, de Jesus RN, Lima AP, Nagasako CK, Fagundes JJ, Ayrizono MDLS, Leal RF. Anti-TNF therapy and immunogenicity in inflammatory bowel diseases: a translational approach. Am J Transl Res 2021; 13:13916-13930. [PMID: 35035733 PMCID: PMC8748125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/09/2021] [Indexed: 06/14/2023]
Abstract
Inflammatory bowel diseases are chronic illnesses that involve intestinal inflammation and are usually diagnosed as Crohn's disease or ulcerative colitis. As these diseases do not have a cure, the goal of treatment is to induce and maintain remission. Monoclonal antibodies have been recognized as the most advanced therapy to avoid complications and reduce the need for surgical approaches. However, although their effectiveness has been proven by several studies, they can trigger the immune system, induce the occurrence of immunogenicity, which may lead to the loss of response and treatment failure. The purpose of this review is to determine what are the main mechanisms involved in IBD; to assess the recommended treatments; to explore the mechanisms of immunogenicity. We also try to explain the detection and describe the existing advances that make possible the clinical application of these approaches.
Collapse
Affiliation(s)
- Lívia Moreira Genaro
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Luís Eduardo Miani Gomes
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Ana Paula Menezes de Freitas Franceschini
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Hugo Dugolin Ceccato
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Rafael Nascimento de Jesus
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Amanda Pereira Lima
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Cristiane Kibune Nagasako
- Department of Gastroenterology, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - João José Fagundes
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| | - Raquel Franco Leal
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Department of Surgery, School of Medical Sciences, University of Campinas (UNICAMP)Campinas, Brazil
| |
Collapse
|
15
|
Riyadh Mohsen E, Ali NH, Aldaoseri HA. Immunologic Parameters for Disease Activity in Rheumatoid Arthritis. ARCHIVES OF RAZI INSTITUTE 2021; 76:1095-1105. [PMID: 35096346 PMCID: PMC8790970 DOI: 10.22092/ari.2021.355758.1718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/09/2021] [Indexed: 05/25/2023]
Abstract
This study aimed to determine the correlation of disease activity of rheumatoid arthritis (RA) with Th17/regulatory T cell (Treg) and Forkhead box protein 3 (Foxp3) cells ratio in patients under therapy with anti-tumor necrosis factor (TNF)-α. Totally, 84 patients with RA and 13 healthy controls were included in this case-control study. The patients were divided into four groups to receive only methotrexate (MTX) (n=25), monotherapy (anti-TNF) (n=18), and combined therapy (MTX+anti-TNF) (n=26); however, one group received no medications (n=15) and was regarded as a positive control. Other 13 healthy controls that were considered negative controls were also enrolled in this study. Patients with RA were attending Basrah General Hospital, Rheumatology Unit, Biological Therapy Center for receiving anti-TNF therapy. Flow cytometry was used for measuring Treg/Foxp3 and Th17 markers, and the DAS-28 score was utilized to measure RA disease activity. Anti-TNF inhibitors (e.g., infliximab and etanercept), as well as other inflammatory and hematological parameters (e.g., erythrocyte sedimentation rate, total white blood cells, lymphocytes, monocytes, and neutrophil counts), were also measured in this study. DAS-28 as a disease activity score was significantly correlated with Th17/Treg/Foxp3 ratio and the Th17 cells count. Statistically, Th17/Treg/Foxp3 ratio was not correlated with body mass index, morning stiffness, and duration of the disease. Th17/Treg/Foxp3 ratio correlated significantly with DAS-28 as an RA disease activity. The lower Treg/Foxp3 frequency led to the higher DAS score reflecting higher disease activity. In the combined therapy group, disease activity was found lower than that in other patient groups indicating the effect of this combination on the relationship between MTX and anti-TNF. This study demonstrated that the main advantage of this combined therapy in RA patients was the reversion of Th17 cell expansion.
Collapse
Affiliation(s)
| | - N H Ali
- College of Medicine, University of Basrah, Basrah, Iraq
| | - H A Aldaoseri
- College of Medicine, University of Basrah, Basrah, Iraq
| |
Collapse
|
16
|
Al-Rahim AM, AlChalabi R, Al-Saffar AZ, Sulaiman GM, Albukhaty S, Belali T, Ahmed EM, Khalil KAA. Folate-methotrexate loaded bovine serum albumin nanoparticles preparation: an in vitro drug targeting cytokines overwhelming expressed immune cells from rheumatoid arthritis patients. Anim Biotechnol 2021; 34:166-182. [PMID: 34319853 DOI: 10.1080/10495398.2021.1951282] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The study planned to estimate biological parameters linked to rheumatoid arthritis (RA) patients, detecting the influence of MTX and biotherapy treatments on these parameters and synthesizing methotrexate bovine serum albumin nanoparticles linked to folate (FA-MTX-BSA NPs) to reduce the overwhelming expression of inflammatory cytokines. Inflammatory parameters showed significant increases in newly diagnosed and MTX-receiving groups while no changes were observed in the biotherapy-maintained group. MTX-loaded BSA nanoparticles were fabricated by the desolvation method and further linked to activated folic acid to obtain FA-MTX-BSA NPs. FA-MTX-BSA NPs were successfully characterized within the nanoscale range using different screening techniques. FA-MTX-BSA NPs showed an in vitro release in a sustained manner. The potential of MTX, MTX-BSA NPs, and FA-MTX-BSA NPs in inducing cytokine level reduction was detected. Significant decreases in interleukin- 1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) levels were obtained in cultures treated with FA-MTX-BSA NPs compared to the untreated culture in a dose-dependent pattern. Furthermore, FA-MTX-BSA NPs comparing with MTX and MTX-BSA NPs exhibited a significant advanced effect in decreasing cytokines levels. Accordingly, the conjunction of BSA NPs and MTX linked to folate potentially reduced cytokines manifestation in RA.
Collapse
Affiliation(s)
- Aya M Al-Rahim
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Rawaa AlChalabi
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Ali Z Al-Saffar
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | | | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan, Iraq
| | - Tareg Belali
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Elsadig M Ahmed
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia.,Department of Clinical Chemistry, Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti, Sudan
| | - Khalil A A Khalil
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Medicine and Health Sciences, University of Hodeidah, Hodeidah, Yemen
| |
Collapse
|
17
|
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
|
18
|
Hoshitsuki K, Rathod S, Ramsey MJ, Zhu L, Moreland LW, Fernandez CA. Adalimumab Immunogenicity Is Negatively Correlated with Anti-Hinge Antibody Levels in Patients with Rheumatoid Arthritis. J Pharmacol Exp Ther 2020; 375:488-497. [PMID: 33008870 PMCID: PMC7718728 DOI: 10.1124/jpet.120.000179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Patients with rheumatoid arthritis (RA) are frequently treated with anti-tumor necrosis factor-α immunoglobulin therapy but develop neutralizing antibodies against these drugs, necessitating therapeutic monitoring of drug concentrations and anti-drug antibodies. Patients with RA have multiple factors related to their autoimmune disposition that interfere with conventionally used methods to detect anti-drug antibodies. Currently deployed analytical methods have significant limitations that hinder clinical interpretation and/or routine use, and no method can detect immunogenicity and drug levels simultaneously to provide clinically meaningful recommendations. Given these limitations, the objective of this study was to identify sources of and associations with assay interference in patients with RA. We designed a modular immunogenicity and drug concentration detection technology to identify the factors that interfere with the detection of adalimumab and anti-adalimumab antibodies in a cohort of 206 patients with RA. Patients were included from the University of Pittsburgh Rheumatoid Arthritis Comparative Effectiveness Research registry. In this cohort, we analyzed clinical and plasma factors associated with anti-adalimumab and anti-hinge antibodies. A novel flow cytometry-based assay was developed and validated that simultaneously measures adalimumab and anti-adalimumab antibody concentrations, overcoming many of the interference factors that are limitations of conventional assays, including anti-fragment crystallizable (Fc) and anti-hinge antibodies. C-reactive protein (P = 0.035), Disease Activity Score-28 (DAS28) score (P = 0.002), and disease activity category (P = 0.009) were significantly associated with anti-adalimumab antibodies but not with anti-hinge antibodies (P > 0.05). Anti-hinge antibodies were inversely associated with drug-neutralizing antibodies (P = 0.002). In patients with RA, anti-hinge antibodies may have a potential protective effect against the development of anti-adalimumab antibodies. SIGNIFICANCE STATEMENT: Using a novel cytometric assay that simultaneously measures drug and anti-drug antibodies, we overcame many interferences that hinder the clinical interpretation of adalimumab immunogenicity testing. Our investigation in patients with RA demonstrated that immunogenicity impaired the pharmacological action of adalimumab via analysis of RA disease severity markers. We also demonstrate that patients with anti-hinge antibodies had lower anti-adalimumab antibody levels and decreased drug neutralization. Our results suggest that anti-hinge antibodies can predict adalimumab immunogenicity before the start of therapy.
Collapse
Affiliation(s)
- Keito Hoshitsuki
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (K.H., S.R., M.J.R., C.A.F.) and Rheumatology and Clinical Immunology (L.Z., L.W.M.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sanjay Rathod
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (K.H., S.R., M.J.R., C.A.F.) and Rheumatology and Clinical Immunology (L.Z., L.W.M.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Manda J Ramsey
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (K.H., S.R., M.J.R., C.A.F.) and Rheumatology and Clinical Immunology (L.Z., L.W.M.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lei Zhu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (K.H., S.R., M.J.R., C.A.F.) and Rheumatology and Clinical Immunology (L.Z., L.W.M.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Larry W Moreland
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (K.H., S.R., M.J.R., C.A.F.) and Rheumatology and Clinical Immunology (L.Z., L.W.M.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christian A Fernandez
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (K.H., S.R., M.J.R., C.A.F.) and Rheumatology and Clinical Immunology (L.Z., L.W.M.), University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
19
|
Kharlamova N, Hermanrud C, Dunn N, Ryner M, Hambardzumyan K, Vivar Pomiano N, Marits P, Gjertsson I, Saevarsdottir S, Pullerits R, Fogdell-Hahn A. Drug Tolerant Anti-drug Antibody Assay for Infliximab Treatment in Clinical Practice Identifies Positive Cases Earlier. Front Immunol 2020; 11:1365. [PMID: 32793189 PMCID: PMC7385065 DOI: 10.3389/fimmu.2020.01365] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/28/2020] [Indexed: 01/14/2023] Open
Abstract
A subgroup of patients treated with infliximab lose response to the treatment and one reason for this is the development of anti-drug antibodies (ADA). If used optimally, measuring drug and ADA level could lead to a more personalized and efficient treatment regime, and enable identification of ADA-positive patients before the underlying disease flares or allergic reactions occur. With the use of a drug-tolerant ADA assay which can detect ADA irrespective of drug levels in the sample, we determined the impact of ADA on treatment failure to infliximab. The aims of this study were to estimate the real-life optimal serum infliximab (sIFX) level and set a clinical threshold value for a drug-tolerant ADA assay. Trough levels of sIFX were measured with ELISA. Free ADA was measured with two drug-sensitive methods (ELISA and a bioassay) and one drug-tolerant method (PandA). Two real-life cohorts treated with infliximab were included; a cross-sectional cohort including patients with inflammatory rheumatic diseases (n = 270) and a prospective cohort of rheumatoid arthritis (RA) patients (n = 73) followed for 1 year. Normal range of sIFX was estimated from the prospective cohort and an arbitrary optimal drug level was set to be between 1 and 6 μg/mL. Using this range, optimal sIFX was found in only 60% (163/270) of the patients in the cross-sectional cohort. These patients had significantly better treatment response than those with a drug level under 1 μg/mL, who had an ADA frequency of 34% (19/56) using the drug-tolerant method. In the prospective cohort, the drug-tolerant assay could identify 34% (53/155 samples) as ADA positive in samples with sIFX level >0.2 μg/mL. ADA were seldom detected in patients with >1 μg/mL sIFX, with three interesting exceptions. A clinically relevant ADA threshold was determined to be >3 RECL as measured with the drug-tolerant assay. In a real-life setting, there was a substantial number of patients with suboptimal drug levels and a proportion of these had ADA. Both too low and too high drug levels correlated with worse disease, but for different reasons. Adding a drug-tolerant assay enabled detection of ADA earlier and regardless of drug level at time of sampling.
Collapse
Affiliation(s)
- Nastya Kharlamova
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| | - Christina Hermanrud
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| | - Nicky Dunn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| | - Malin Ryner
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| | - Karen Hambardzumyan
- Rheumatology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Nancy Vivar Pomiano
- Rheumatology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Per Marits
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Saedis Saevarsdottir
- Rheumatology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
20
|
Gress K, Bass JA, Funk RS, Morrow RP, Hasenkamp R, Shakhnovich V. Facing Real-World Challenges of Immunogenicity in Pediatric Inflammatory Bowel Disease. Front Immunol 2020; 11:1148. [PMID: 32582213 PMCID: PMC7295975 DOI: 10.3389/fimmu.2020.01148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/11/2020] [Indexed: 01/15/2023] Open
Affiliation(s)
- Kyle Gress
- Georgetown University School of Medicine, Washington, DC, United States
| | - Julie A Bass
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States.,Children's Mercy Kansas City, Kansas City, MO, United States
| | - Ryan S Funk
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ryan P Morrow
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States.,Children's Mercy Kansas City, Kansas City, MO, United States
| | | | - Valentina Shakhnovich
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States.,Children's Mercy Kansas City, Kansas City, MO, United States
| |
Collapse
|
21
|
Nabhan M, Pallardy M, Turbica I. Immunogenicity of Bioproducts: Cellular Models to Evaluate the Impact of Therapeutic Antibody Aggregates. Front Immunol 2020; 11:725. [PMID: 32431697 PMCID: PMC7214678 DOI: 10.3389/fimmu.2020.00725] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022] Open
Abstract
Patients treated with bioproducts (BPs) frequently develop anti-drug antibodies (ADAs) with potential neutralizing capacities leading to loss of clinical response or potential hypersensitivity reactions. Many factors can influence BP immunogenicity and could be related to the patient, the treatment, as well as to the product itself. Among these latter factors, it is now well accepted that BP aggregation is associated with an increased potential for immunogenicity, as aggregates seem to be correlated with ADA development. Moreover, the presence of high-affinity ADAs suggests a CD4 T-cell dependent adaptive immune response and therefore a pivotal role for antigen-presenting cells (APCs), such as dendritic cells (DCs). In this review, we address the in vitro methods developed to evaluate how monoclonal antibodies could trigger the immunization process by focusing on the role of aggregated antibodies in the establishment of this response. In particular, we will present the different cell-based assays that have been used to assess the potential of antibodies and their aggregates to modulate cellular mechanisms leading to activation and the biological parameters (cellular activation markers, proliferation and secreted molecules) that can be measured to evaluate the different cell activation stages and their consequences in the propagation of the immune response. Indeed, the use of such strategies could help evaluate the risk of BP immunogenicity and their role in mitigating this risk.
Collapse
Affiliation(s)
- Myriam Nabhan
- Inserm, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| | - Marc Pallardy
- Inserm, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| | - Isabelle Turbica
- Inserm, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
22
|
Bodio C, Grossi C, Pregnolato F, Favalli EG, Biggioggero M, Marchesoni A, Murgo A, Filippini M, Migliorini P, Caporali R, Pellerito R, Ciccia F, Sarzi-Puttini P, Perosa F, Paolazzi G, Hollan I, Bendtzen K, Meroni PL, Borghi MO. Personalized medicine in rheumatoid arthritis: How immunogenicity impacts use of TNF inhibitors. Autoimmun Rev 2020; 19:102509. [DOI: 10.1016/j.autrev.2020.102509] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 01/30/2023]
|
23
|
Schuster L, Sauter M, Uhl P, Meid A, Haefeli WE, Weiss J, Theile D. Reporter cell assay-based functional quantification of TNF-α-antagonists in serum – a proof-of-principle study for adalimumab. Anal Biochem 2020; 596:113646. [DOI: 10.1016/j.ab.2020.113646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 10/24/2022]
|
24
|
Berends SE, Strik AS, Löwenberg M, D'Haens GR, Mathôt RAA. Clinical Pharmacokinetic and Pharmacodynamic Considerations in the Treatment of Ulcerative Colitis. Clin Pharmacokinet 2020; 58:15-37. [PMID: 29752633 PMCID: PMC6326086 DOI: 10.1007/s40262-018-0676-z] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) of unknown etiology, probably caused by a combination of genetic and environmental factors. The treatment of patients with active UC depends on the severity, localization and history of IBD medication. According to the classic step-up approach, treatment with 5-aminosalicylic acid compounds is the first step in the treatment of mild to moderately active UC. Corticosteroids, such as prednisolone are used in UC patients with moderate to severe disease activity, but only for remission induction therapy because of side effects associated with long-term use. Thiopurines are the next step in the treatment of active UC but monotherapy during induction therapy in UC patients is not preferred because of their slow onset. Therapeutic drug monitoring (TDM) of the pharmacologically active metabolites of thiopurines, 6-thioguanine nucleotide (6-TGN), has proven to be beneficial. Thiopurine S-methyltransferase (TMPT) plays a role in the metabolic conversion pathway of thiopurines and exhibits genetic polymorphism; however, the clinical benefit and relevance of TPMT genotyping is not well established. In patients with severely active UC refractory to corticosteroids, calcineurin inhibitors such as ciclosporin A (CsA) and tacrolimus are potential therapeutic options. These agents usually have a rather rapid onset of action. Monoclonal antibodies (anti-tumor necrosis factor [TNF] agents, vedolizumab) are the last pharmacotherapeutic option for UC patients before surgery becomes inevitable. Body weight, albumin status and antidrug antibodies contribute to the variability in the pharmacokinetics of anti-TNF agents. Additionally, the use of concomitant immunomodulators (thiopurines/methotrexate) lowers the rate of immunogenicity, and therefore the concomitant use of anti-TNF therapy with an immunomodulator may confer some advantage compared with monotherapy in certain patients. TDM of anti-TNF agents could be beneficial in patients with primary nonresponse and secondary loss of response. The potential benefit of applying TDM during vedolizumab treatment has yet to be determined.
Collapse
Affiliation(s)
- Sophie E Berends
- Department Hospital Pharmacy, Academic Medical Center, Amsterdam, The Netherlands.
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| | - Anne S Strik
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Geert R D'Haens
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Ron A A Mathôt
- Department Hospital Pharmacy, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
25
|
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
|
26
|
In Vitro ELISA and Cell-Based Assays Confirm the Low Immunogenicity of VNAR Therapeutic Constructs in a Mouse Model of Human RA: An Encouraging Milestone to Further Clinical Drug Development. J Immunol Res 2020; 2020:7283239. [PMID: 32090129 PMCID: PMC7023846 DOI: 10.1155/2020/7283239] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/19/2019] [Accepted: 01/11/2020] [Indexed: 12/17/2022] Open
Abstract
Anti-drug antibodies (ADAs), specific for biotherapeutic drugs, are associated with reduced serum drug levels and compromised therapeutic response. The impact of ADA on the bioavailability and clinical efficacy of blockbuster anti-hTNF-α monoclonal antibodies is well recognised, especially for adalimumab and infliximab treatments, with the large and complex molecular architecture of classical immunoglobulin antibody drugs, in part, responsible for the immunogenicity seen in patients. The initial aim of this study was to develop solid-phase enzyme-linked immunosorbent assays (ELISA) and an in vitro cell-based method to accurately detect ADA and estimate its impact on the preclinical in vivo efficacy outcomes of two novel, nonimmunoglobulin VNAR fusion anti-hTNF-α biologics (Quad-X™ and D1-NDure™-C4) and Humira®, a brand of adalimumab. Serum drug levels and the presence of ADA were determined in a transgenic mouse model of polyarthritis (Tg197) when Quad-X™ and Humira® were dosed at 1 mg/kg and D1-NDure™-C4 was dosed at 30 mg/kg. The serum levels of the Quad-X™ and D1-NDure™-C4 modalities were consistently high and comparable across all mice within the same treatment groups. In 1 mg/kg and 3 mg/kg Quad-X™- and 30 mg/kg D1-NDure™-C4-treated mice, an average trough drug serum concentration of 8 μg/mL, 50 μg/mL, and 350 μg/mL, respectively, were estimated. In stark contrast, Humira® trough serum concentrations in the 1 mg/kg treatment group ranged from <0.008 μg/mL to 4 μg/mL with trace levels detected in 7 of the 8 animals treated. Trough serum Humira® and Quad-X™ concentrations in 3 mg/kg treatment samples were comparable; however, the functionality of the detected Humira® serum was significantly compromised due to neutralising ADA. The impact of ADA went beyond the simple and rapid clearance of Humira®, as 7/8 serum samples also showed no detectable capacity to neutralise hTNF-α-mediated cytotoxicity in a murine fibrosarcoma (L929) cell assay. The neutralisation capacity of all the VNAR constructs remained unchanged at the end of the experimental period (10 weeks). The data presented in this manuscript goes some way to explain the exciting outcomes of the previously published preclinical in vivo efficacy data, which showed complete control of disease at Quad-X™ concentrations of 0.5 mg/kg, equivalent to 10x the in vivo potency of Humira®. This independent corroboration also validates the robustness and reliability of the assay techniques reported in this current manuscript, and while it comes with the caveat of a mouse study, it does appear to suggest that these particular VNAR constructs, at least, are of low inherent immunogenicity.
Collapse
|
27
|
Vaisman-Mentesh A, Rosenstein S, Yavzori M, Dror Y, Fudim E, Ungar B, Kopylov U, Picard O, Kigel A, Ben-Horin S, Benhar I, Wine Y. Molecular Landscape of Anti-Drug Antibodies Reveals the Mechanism of the Immune Response Following Treatment With TNFα Antagonists. Front Immunol 2019; 10:2921. [PMID: 31921180 PMCID: PMC6930160 DOI: 10.3389/fimmu.2019.02921] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/28/2019] [Indexed: 12/25/2022] Open
Abstract
Drugs formulated from monoclonal antibodies (mAbs) are clinically effective in various diseases. Repeated administration of mAbs, however, elicits an immune response in the form of anti-drug-antibodies (ADA), thereby reducing the drug's efficacy. Notwithstanding their importance, the molecular landscape of ADA and the mechanisms involved in their formation are not fully understood. Using a newly developed quantitative bio-immunoassay, we found that ADA concentrations specific to TNFα antagonists can exceed extreme concentrations of 1 mg/ml with a wide range of neutralization capacity. Our data further suggest a preferential use of the λ light chain in a subset of neutralizing ADA. Moreover, we show that administration of TNFα antagonists result in a vaccine-like response whereby ADA formation is governed by the extrafollicular T cell-independent immune response. Our bio-immunoassay coupled with insights on the nature of the immune response can be leveraged to improve mAb immunogenicity assessment and facilitate improvement in therapeutic intervention strategies.
Collapse
Affiliation(s)
- Anna Vaisman-Mentesh
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Shai Rosenstein
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Miri Yavzori
- Gastroenterology Department, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Yael Dror
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Ella Fudim
- Gastroenterology Department, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Bella Ungar
- Gastroenterology Department, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Uri Kopylov
- Gastroenterology Department, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Orit Picard
- Gastroenterology Department, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Aya Kigel
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Shomron Ben-Horin
- Gastroenterology Department, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Itai Benhar
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Yariv Wine
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
28
|
von Bremen H, Gamboa LM, Zernicke J, Poddubnyy D, Feist E. Evaluation von Immunoassays zum Nachweis von Anti-Drug Antikörpern gegen Infliximab und korrespondierenden Infliximab Serumspiegeln bei Spondyloarthritis im klinischen Alltag. AKTUEL RHEUMATOL 2019. [DOI: 10.1055/a-1033-8153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Zusammenfassung
Hintergrund TNF-α-Inhibitoren sind wirksame Medikamente bei vielen chronisch-inflammatorischen Erkrankungen. Trotz eines akzeptablen Wirkansprechens gibt es einen beachtlichen Anteil an Patienten, bei denen ein primärer oder sekundärer Wirkverlust beobachtet wird. Eine mögliche Erklärung für das fehlende Therapieansprechen wird in der Immunogenität von Biologika gesehen. Die resultierenden Anti-Drug-Antikörper (ADA) können die Wirkung der Biologika abschwächen und/oder unerwünschte Arzneimittelwirkungen hervorrufen.
Ziel Durch diese Arbeit sollten 2 neue Testverfahren zum Nachweis von ADA gegen Infliximab und die korrespondierenden Serumspiegel evaluiert werden. Dazu gehört ebenfalls die Klärung der Praktikabilität eines therapeutischen Drugmonitorings (TDM) sowie das Erfassen von ADA assoziierten Nebenwirkungen und Therapieversagen für den klinischen Alltag. Es sollte eine Einschätzung darüber gegeben werden, welche Effekte das Auftreten von ADA auf den Medikamentenspiegel und den Verlauf von entzündlich-rheumatischen Erkrankungen im Rahmen der Verlaufsuntersuchungen hat.
Patienten und Methodik Die vorliegende Studie umfasst 30 mit Infliximab behandelte Patienten (30% weiblich, mittleres Alter 55,5 Jahre, min. 33 Jahre, max. 73 Jahre) mit zuvor gesicherter PsA (20%) oder axialer SpA (80%). Bei 36,7% bestand eine Begleitmedikation mit einem konventionellen Basistherapeutikum (cDMARD). Bei den vorhandenen Proben wurden zu 2 Zeitpunkten (ZP) der Serumspiegel des Therapieantikörpers, sowie die totalen ADA (tADA) und freien ADA (fADA) mittels Enzyme-Linked-Immuno-Sorbent-Assay (ELISA) bestimmt und in Zusammenhang mit der Krankheitsaktivität gesetzt. Weiter wurde die Inter-Assay-Variabilität untersucht.
Ergebnisse Bei 19 Patienten (63,3%) wurden positive tADA nachgewiesen, bei 13 Patienten (43,3%) auch fADA. Bei Nachweis von positiven tADA fanden sich zu 94,7% zeitweise subtherapeutische Infliximabkonzentrationen. Hohe tADA Titer gingen mit einer höheren Krankheitsaktivität einher. fADA traten nur bei mittleren bis hohen tADA-Titern auf. Bei 30% der Patienten fanden sich tADA trotz klinischer Remission der Erkrankung. Zwischen den beiden Assays zur Erfassung des Infliximab Serumspiegels gab es zu beiden Messzeitpunkten eine hohe Interassay-Übereinstimmung (1. ZP r=0,78, p=0,01; 2. ZP r=0,96, p=0,01).
Schlussfolgerungen Eine Erfassung von ADA sollte im Kontext mit den korrespondierenden Medikamenten-Serumspiegeln im Rahmen der Verlaufsuntersuchungen erfolgen, um relevante Informationen für das Monitoring einer Infliximabtherapie im klinischen Alltag zu erhalten. Bei der Untersuchung auf ADA sollte zwischen niedrigen und hohen tADA unterschieden werden, da hohe Spiegel mit dem Auftreten von fADA und erhöhter Krankheitsaktivität assoziiert sind. Die Ergebnisse müssen jedoch immer auf den individuellen Fall im Zusammenhang mit der klinischen Situation bezogen werden, da ADA sogar bei klinischer Remission auftreten können.
Collapse
Affiliation(s)
- Helene von Bremen
- Klinik für Rheumatologie und Klinische Immunologie, Charité Universitätsmedizin Berlin Campus Charité Mitte, Berlin
| | | | - Jan Zernicke
- Klinik für Rheumatologie und Klinische Immunologie, Charité Universitätsmedizin Berlin Campus Charité Mitte, Berlin
| | - Denis Poddubnyy
- Rheumatologie, Med. Klinik I, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Berlin
| | - Eugen Feist
- Klinik für Rheumatologie und Klinische Immunologie, Charité Universitätsmedizin Berlin Campus Charité Mitte, Berlin
- Helios Fachklinik für Rheumatologie, Vogelsang-Gommern
| |
Collapse
|
29
|
Ogrič M, Žigon P, Drobne D, Štabuc B, Sodin-Semrl S, Čučnik S, Praprotnik S. Routinely utilized in-house assays for infliximab, adalimumab and their anti-drug antibody levels. Immunol Res 2019; 66:726-736. [PMID: 30569379 DOI: 10.1007/s12026-018-9050-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
By monitoring serum concentrations of infliximab (IFX) and adalimumab (ADL) and levels of their antibodies in patients with inflammatory diseases, clinicians can adjust dose and increase safety and effectiveness of treatment. The aim was to develop and validate in-house enzyme-linked immunosorbent assays (ELISAs) for IFX and ADL, together with anti-IFX and anti-ADL ELISAs for routine detection and further analysis with acid dissociation of immune complexes. Furthermore, the objectives were to compare in-house assays with commercial ELISAs and reporter gene assays (RGAs) and to determine cross-reactivity between original Remicade®/Remsima™ and their antibodies. In-house ELISAs were validated (imprecision, accuracy among other criteria) and compared with commercial apDia ELISAs and RGAs. Correlation coefficients, intraclass correlation coefficients, agreement, and bias were calculated. All in-house ELISAs gave precise and repeatable results. The immune complexes between IFX and anti-IFX were found in 3% of samples, while complexes between ADL and anti-ADL were found in 14% of samples. Significant correlations were found between in-house and apDia ELISAs and RGA for IFX, ADL, anti-IFX, and anti-ADL results. Remicade®, Remsima™, and their antibodies could be accurately measured with either apDia or in-house IFX and anti-IFX ELISAs. Accurate and precise in-house ELISAs, highly comparable with commercial ELISAs and RGAs, were developed and validated for routine analysis of samples of patients treated with IFX (Remicade® or Remsima™) or ADL providing substantial cost benefit. Complex dissociation identified samples with anti-IFX-IFX (3%) and anti-ADL-ADL (14%) complexes indicating the benefit of adding acid dissociation to therapeutic drug monitoring of IFX and ADL.
Collapse
Affiliation(s)
- Manca Ogrič
- Department of Rheumatology, University Medical Centre Ljubljana, SI-1000, Ljubljana, Slovenia.,Faculty of Pharmacy, Chair of Clinical Biochemistry, University of Ljubljana, SI-1000, Ljubljana, Slovenia
| | - Polona Žigon
- Department of Rheumatology, University Medical Centre Ljubljana, SI-1000, Ljubljana, Slovenia
| | - David Drobne
- Department of Gastroenterology, University Medical Centre Ljubljana, SI-1000, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, SI-1000, Ljubljana, Slovenia
| | - Borut Štabuc
- Department of Gastroenterology, University Medical Centre Ljubljana, SI-1000, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, SI-1000, Ljubljana, Slovenia
| | - Snezna Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, SI-1000, Ljubljana, Slovenia.,Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, SI-6000, Koper, Slovenia
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, SI-1000, Ljubljana, Slovenia. .,Faculty of Pharmacy, Chair of Clinical Biochemistry, University of Ljubljana, SI-1000, Ljubljana, Slovenia.
| | - Sonja Praprotnik
- Department of Rheumatology, University Medical Centre Ljubljana, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
30
|
Modelling of the Time-Varying Pharmacokinetics of Therapeutic Monoclonal Antibodies: A Literature Review. Clin Pharmacokinet 2019; 59:37-49. [DOI: 10.1007/s40262-019-00816-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
31
|
Real-Fernández F, Pregnolato F, Cimaz R, Papini AM, Borghi MO, Meroni PL, Rovero P. Detection of anti-adalimumab antibodies in a RA responsive cohort of patients using three different techniques. Anal Biochem 2019; 566:133-138. [DOI: 10.1016/j.ab.2018.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/27/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022]
|
32
|
Discrimination of Anti-drug Antibodies With Neutralizing Capacity in Infliximab- and Adalimumab-Treated Patients: Comparison of the Homogeneous Mobility Shift Assay and the Affinity Capture and Elution Assay. Ther Drug Monit 2018; 40:705-715. [DOI: 10.1097/ftd.0000000000000553] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
33
|
Goncalves J, Santos M, Acurcio R, Iria I, Gouveia L, Matos Brito P, Catarina Cunha-Santos A, Barbas A, Galvão J, Barbosa I, Aires da Silva F, Alcobia A, Cavaco M, Cardoso M, Delgado Alves J, Carey JJ, Dörner T, Eurico Fonseca J, Palmela C, Torres J, Lima Vieira C, Trabuco D, Fiorino G, Strik A, Yavzori M, Rosa I, Correia L, Magro F, D'Haens G, Ben-Horin S, Lakatos PL, Danese S. Antigenic response to CT-P13 and infliximab originator in inflammatory bowel disease patients shows similar epitope recognition. Aliment Pharmacol Ther 2018; 48:507-522. [PMID: 29873091 DOI: 10.1111/apt.14808] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/18/2018] [Accepted: 04/25/2018] [Indexed: 12/14/2022]
Abstract
AIM To test the cross-immunogenicity of anti-CT-P13 IBD patients' sera to CT-P13/infliximab originator and the comparative antigenicity evoked by CT-P13/infliximab originator sera. METHODS Sera of patients with IBD with measurable anti-CT-P13 antibodies were tested for their cross-reactivity to 5 batches of infliximab originator and CT-P13. Anti-drug antibody positive sera from treated patients were used to compare antigenic epitopes. RESULTS All 42 anti-CT-P13 and 37 anti-infliximab originator IBD sera were cross-reactive with infliximab originator and CT-P13 respectively. Concentration of anti-drug antibodies against infliximab originator or CT-P13 were strongly correlated both for IgG1 and IgG4 (P < 0.001). Anti-CT-P13 sera of patients with IBD (n = 32) exerted similar functional inhibition on CT-P13 or infliximab originator TNF binding capacity and showed reduced binding to CT-P13 in the presence of five different batches of CT-P13 and infliximab originator. Anti-CT-P13 and anti-infliximab originator IBD sera selectively enriched phage-peptides from the VH (CDR1 and CDR3) and VL domains (CDR2 and CDR3) of infliximab. Sera reactivity detected major infliximab epitopes in these regions of infliximab in 60%-79% of patients, and no significant differences were identified between CT-P13 and infliximab originator immunogenic sera. Minor epitopes were localised in framework regions of infliximab with reduced antibody reactivity shown, in 30%-50% of patients. Monoclonal antibodies derived from naïve individuals and ADA-positive IBD patients treated with CT-P13 provided comparable epitope specificity to five different batches of CT-P13 and infliximab originator. CONCLUSIONS These results strongly support a similar antigenic profile for infliximab originator and CT-P13, and point toward a safe switching between the two drugs in anti-drug antibody negative patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - A Barbas
- Oeiras, Portugal
- Carnaxide, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ovacik M, Lin K. Tutorial on Monoclonal Antibody Pharmacokinetics and Its Considerations in Early Development. Clin Transl Sci 2018; 11:540-552. [PMID: 29877608 PMCID: PMC6226118 DOI: 10.1111/cts.12567] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/11/2018] [Indexed: 12/21/2022] Open
Abstract
The tutorial introduces the readers to the fundamentals of antibody pharmacokinetics (PK) in the context of drug development. Topics covered include an overview of antibody development, PK characteristics, and the application of antibody PK/pharmacodynamics (PD) in research and development decision-making. We also discuss the general considerations for planning a nonclinical PK program and describe the types of PK studies that should be performed during early development of monoclonal antibodies.
Collapse
Affiliation(s)
- Meric Ovacik
- Department of Preclinical and Translational Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Kedan Lin
- Clinical Pharmacology, NGM Biopharmaceuticals, Inc., South San Francisco, California, USA
| |
Collapse
|
35
|
Naviglio S, Giuffrida P, Stocco G, Lenti MV, Ventura A, Corazza GR, Di Sabatino A. How to predict response to anti-tumour necrosis factor agents in inflammatory bowel disease. Expert Rev Gastroenterol Hepatol 2018; 12:797-810. [PMID: 29957083 DOI: 10.1080/17474124.2018.1494573] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anti-tumor necrosis factor (TNF) agents have changed the therapeutic approach to inflammatory bowel disease (IBD). However, a considerable proportion of patients either do not primarily respond or lose response to treatment. Despite the long-standing experience in the use of these drugs, still there is the need of identifying the possible predictors of efficacy. Areas covered: We critically review the current knowledge on predictors of response to anti-TNF therapy - both those available in clinical practice and those still under investigation. Multiple factors are involved in treatment success, including disease phenotype and severity, adherence to medications, and pharmacogenomic, pharmacokinetic, and immunologic factors. Literature search was conducted in PubMed using keywords 'inflammatory bowel disease,' 'Crohn's disease,' and 'ulcerative colitis,' matched with 'antitumor necrosis factor,' 'biologic therapy,' 'clinical response,' 'predictors,' and 'efficacy,' Relevant articles were selected for review. Expert commentary: While the role of several factors in clinical practice is clearly established, other investigational markers have been proposed, mostly in small studies, yet for many of them little external validation exists. Therapeutic drug monitoring is emerging as a pivotal strategy to guide decisions in clinical practice. In the near future, novel markers could improve our ability to direct treatment and personalize therapy.
Collapse
Affiliation(s)
- Samuele Naviglio
- a Institute for Maternal and Child Health IRCCS Burlo Garofolo , Trieste , Italy
- b Department of Medicine, Surgery, and Health Sciences , University of Trieste , Trieste , Italy
| | - Paolo Giuffrida
- c First Department of Internal Medicine, San Matteo Hospital Foundation , University of Pavia , Pavia , Italy
| | - Gabriele Stocco
- d Department of Life Science , University of Trieste , Trieste , Italy
| | - Marco Vincenzo Lenti
- c First Department of Internal Medicine, San Matteo Hospital Foundation , University of Pavia , Pavia , Italy
| | - Alessandro Ventura
- a Institute for Maternal and Child Health IRCCS Burlo Garofolo , Trieste , Italy
- b Department of Medicine, Surgery, and Health Sciences , University of Trieste , Trieste , Italy
| | - Gino Roberto Corazza
- c First Department of Internal Medicine, San Matteo Hospital Foundation , University of Pavia , Pavia , Italy
| | - Antonio Di Sabatino
- c First Department of Internal Medicine, San Matteo Hospital Foundation , University of Pavia , Pavia , Italy
| |
Collapse
|
36
|
Immunogenicity of Therapeutic Antibodies: Monitoring Antidrug Antibodies in a Clinical Context. Ther Drug Monit 2018; 39:327-332. [PMID: 28463887 DOI: 10.1097/ftd.0000000000000404] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One of the factors that may impact drug levels of therapeutic antibodies in patients is immunogenicity, with potential loss of efficacy. Nowadays, many immunogenicity assays are available for testing antidrug antibodies (ADA). In this article, we discuss different types of immunogenicity assays and their clinical relevance in terms of drug tolerance, relation with pharmacokinetics (PK), neutralizing antibodies, potential adverse events associated with ADA, and prediction of ADA production. Drug-tolerant assays can provide insight into the process of immunogenicity, but for clinical management, these assays do not necessarily outperform drug-sensitive assays. The usefulness of any ADA assay for clinical decision making will be larger when drug concentrations are also measured, and this is true, in particular, for drug-tolerant assays.
Collapse
|
37
|
Carman N, Mack DR, Benchimol EI. Therapeutic Drug Monitoring in Pediatric Inflammatory Bowel Disease. Curr Gastroenterol Rep 2018; 20:18. [PMID: 29623442 DOI: 10.1007/s11894-018-0623-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
PURPOSE OF REVIEW Therapeutic drug monitoring (TDM) has emerged as a useful tool to optimize the use of drug therapies in adults with inflammatory bowel disease (IBD), including both Crohn's disease (CD) and ulcerative colitis (UC), especially during the use of biological therapies, for which the pharmacokinetics and pharmacodynamics are highly variable among patients. Fewer data exist in children. This review examines the current literature on TDM in pediatric IBD. RECENT FINDINGS Drug clearance is affected by a number of patient and disease factors. For thiopurines, adjusting dosing by monitoring 6-thioguanine (6TGN) and 6-methylmercaptopurine ((6MMP) levels is demonstrated to maximize response and minimize toxicity, while monitoring metabolite levels when treating with anti-tumor necrosis factor (anti-TNF) remain controversial. While in adults the use of TDM in the setting of loss of response to anti-TNF therapy is established, in children, only a small number of studies exist, but these too have encouraging results. There are however, conflicting data regarding the optimal timing of TDM, comparing "reactive" monitoring and "proactive" monitoring. No such data exist in pediatrics. TDM is cost-effective, and dose reduction may represent a safety benefit. There are limited adult data for use of TDM for the newer biologics, vedolizumab and ustekinumab, but early results suggest similarly promising utility. The use of TDM in pediatric IBD is increasing in clinical practice, with similar efficacy to adults demonstrated in children with loss of response to anti-TNF therapy. More prospective studies are needed in children to examine proactive monitoring and utility of TDM with newer biologics.
Collapse
Affiliation(s)
- Nicholas Carman
- CHEO Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada.
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada.
| | - David R Mack
- CHEO Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Eric I Benchimol
- CHEO Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
38
|
Ju S, Lee S, Chakkarapani SK, Kim K, Yu H, Kang SH. One-Shot Dual-Code Immunotargeting for Ultra-Sensitive Tumor Necrosis Factor-α Nanosensors by 3D Enhanced Dark-Field Super-Resolution Microscopy. Anal Chem 2018; 90:5100-5107. [DOI: 10.1021/acs.analchem.7b05107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Soyeong Ju
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Seungah Lee
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Suresh Kumar Chakkarapani
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Kyungsoo Kim
- Department of Applied Mathematics, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Hyunung Yu
- Nanobio Fusion Research Center, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Seong Ho Kang
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| |
Collapse
|
39
|
van Rosmalen M, Ni Y, Vervoort DFM, Arts R, Ludwig SKJ, Merkx M. Dual-Color Bioluminescent Sensor Proteins for Therapeutic Drug Monitoring of Antitumor Antibodies. Anal Chem 2018; 90:3592-3599. [PMID: 29443503 PMCID: PMC5843950 DOI: 10.1021/acs.analchem.8b00041] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Monitoring the levels of therapeutic antibodies in individual patients would allow patient-specific dose optimization, with the potential for major therapeutic and financial benefits. Our group recently developed a new platform of bioluminescent sensor proteins (LUMABS; LUMinescent AntiBody Sensor) that allow antibody detection directly in blood plasma. In this study, we targeted four clinically important therapeutic antibodies, the Her2-receptor targeting trastuzumab, the anti-CD20 antibodies rituximab and obinutuzumab, and the EGFR-blocking cetuximab. A strong correlation was found between the affinity of the antibody binding peptide and sensor performance. LUMABS sensors with physiologically relevant affinities and decent sensor responses were obtained for trastuzumab and cetuximab using mimotope and meditope peptides, respectively, with affinities in the 10-7 M range. The lower affinity of the CD20-derived cyclic peptide employed in the anti-CD20 LUMABS sensor ( Kd = 10-5 M), translated in a LUMABS sensor with a strongly attenuated sensor response. The trastuzumab and cetuximab sensors were further characterized with respect to binding kinetics and their performance in undiluted blood plasma. For both antibodies, LUMABS-based detection directly in plasma compared well to the analytical performance of commercial ELISA kits. Besides identifying important design parameters for the development of new LUMABS sensors, this work demonstrates the potential of the LUMABS platform for point-of-care detection of therapeutic antibodies.
Collapse
Affiliation(s)
- Martijn van Rosmalen
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Yan Ni
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Daan F M Vervoort
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Remco Arts
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Susann K J Ludwig
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| | - Maarten Merkx
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems (ICMS), Department of Biomedical Engineering , Eindhoven University of Technology , P.O. Box 513, 5600 MB Eindhoven , The Netherlands
| |
Collapse
|
40
|
van Hoeve K, Hoffman I, Vermeire S. Therapeutic drug monitoring of anti-TNF therapy in children with inflammatory bowel disease. Expert Opin Drug Saf 2017; 17:185-196. [PMID: 29202588 DOI: 10.1080/14740338.2018.1413090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Karen van Hoeve
- Department of Pediatric Gastroenterology, University Hospitals Leuven, Leuven, Belgium
| | - Ilse Hoffman
- Department of Pediatric Gastroenterology, University Hospitals Leuven, Leuven, Belgium
| | - Severine Vermeire
- Department of Gastroenterology & Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Prado MS, Bendtzen K, Andrade LEC. Biological anti-TNF drugs: immunogenicity underlying treatment failure and adverse events. Expert Opin Drug Metab Toxicol 2017; 13:985-995. [DOI: 10.1080/17425255.2017.1360280] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Mônica Simon Prado
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Klaus Bendtzen
- Institute for Inflammation Research, Rigshospitalet University Hospital, Copenhagen, Denmark
| | | |
Collapse
|
42
|
Morera Y, Sánchez J, Bequet-Romero M, Selman-Housein KH, de la Torre A, Hernández-Bernal F, Martín Y, Garabito A, Piñero J, Bermúdez C, de la Torre J, Ayala M, Gavilondo JV. Specific humoral and cellular immune responses in cancer patients undergoing chronic immunization with a VEGF-based therapeutic vaccine. Vaccine 2017; 35:3582-3590. [DOI: 10.1016/j.vaccine.2017.05.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 05/02/2017] [Accepted: 05/07/2017] [Indexed: 12/26/2022]
|
43
|
Hindryckx P, Novak G, Vande Casteele N, Khanna R, Laukens D, Jairath V, Feagan BG. Incidence, Prevention and Management of Anti-Drug Antibodies Against Therapeutic Antibodies in Inflammatory Bowel Disease: A Practical Overview. Drugs 2017; 77:363-377. [PMID: 28233275 DOI: 10.1007/s40265-017-0693-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The introduction of biologic therapy has revolutionized the treatment of inflammatory bowel disease (IBD). However, like all therapeutic proteins, monoclonal antibodies have immunogenic potential which is influenced by multiple drug- and patient-related factors. The reported incidence of anti-drug antibodies (ADAs) towards biologic drugs in IBD varies greatly in the literature and depends not only on differences in sensitization but also on the assay methodology and the timepoint of measurement. Sensitization with formation of ADAs is associated with an increased risk of infusion reactions, accelerated drug clearance, and a loss of response (LOR) to drug. Recently, a greater understanding of the pharmacokinetics of therapeutic antibodies has led to the development of new strategies to reduce immunogenicity and more efficient use of these drugs. These preventive strategies include regular scheduled dosing with maintenance of stable therapeutic trough drug concentrations, and co-administration of an immunosuppressive. Sub-therapeutic drug concentrations with low levels of ADAs can generally be overcome with dose escalation, whereas the presence of high concentrations of ADAs requires a switch to another therapeutic agent.
Collapse
Affiliation(s)
- Pieter Hindryckx
- Robarts Clinical Trials, University of Western Ontario, London, ON, Canada.,Department of Gastroenterology, University of Ghent, Ghent, Belgium
| | - Gregor Novak
- Robarts Clinical Trials, University of Western Ontario, London, ON, Canada.,Department of Gastroenterology, Ljubljana University Medical Centre, Ljubljana, Slovenia
| | - Niels Vande Casteele
- Robarts Clinical Trials, University of Western Ontario, London, ON, Canada.,Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Reena Khanna
- Robarts Clinical Trials, University of Western Ontario, London, ON, Canada.,Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Debby Laukens
- Department of Gastroenterology, University of Ghent, Ghent, Belgium
| | - Vipul Jairath
- Robarts Clinical Trials, University of Western Ontario, London, ON, Canada.,Department of Medicine, University of Western Ontario, London, ON, Canada.,Department of Epidemiology and Biostatistics, University of Western Ontario, London, ON, Canada
| | - Brian G Feagan
- Robarts Clinical Trials, University of Western Ontario, London, ON, Canada. .,Department of Medicine, University of Western Ontario, London, ON, Canada. .,Department of Epidemiology and Biostatistics, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
44
|
Bendtzen K, Steenholdt C. Letter: can addition of an immunomodulator really reverse antibody formation and loss of response in patients treated with adalimumab? Aliment Pharmacol Ther 2017; 45:759-760. [PMID: 28150453 DOI: 10.1111/apt.13921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- K Bendtzen
- Institute for Inflammation Research, Rigshospitalet, Copenhagen, Denmark
| | - C Steenholdt
- Department of Gastroenterology, Herlev Hospital, Herlev, Denmark
| |
Collapse
|
45
|
Bian S, Ferrante M, Gils A. Validation of a Drug-Resistant Anti-Adalimumab Antibody Assay to Monitor Immunogenicity in the Presence of High Concentrations of Adalimumab. AAPS JOURNAL 2016; 19:468-474. [PMID: 27873119 DOI: 10.1208/s12248-016-0018-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/14/2016] [Indexed: 11/30/2022]
Abstract
With respect to patient safety and long-term efficacy, immunogenicity of therapeutic antibodies remains an important issue. Pre-treatment of samples using either higher temperature or acidification in order to separate drug/anti-drug antibody complexes has been implemented in the traditional bridging assay and an in-house-developed affinity capture elution assay but only a limited drug tolerance was obtained. In this study, we aim to apply a drug-resistant anti-drug antibody assay to adalimumab through a combination of adalimumab/anti-adalimumab antibody complex precipitation and acid dissociation. A linear dose-response curve ranging from 3.1 to 200 ng/mL was obtained in 1/125 diluted serum, allowing detection of anti-adalimumab antibody concentrations up to 25 μg/mL equivalents MA-ADM6A10, a calibrator anti-adalimumab antibody. The cut-off point for detection was determined using 16 samples of adalimumab naïve patients and set at 0.39 μg/mL equivalents. Validation of the assay revealed that no detectable anti-adalimumab antibody concentrations were found in samples with either a positive anti-infliximab antibody concentration, a physiologic concentration of TNFα, or a high concentration of rheumatoid factor. Full recoveries were obtained when various concentrations of adalimumab (0, 1, 10, and 50 μg/mL) were spiked to 1, 2, and 4 μg/mL of MA-ADM6A10. Spiking of 50 μg/mL adalimumab to eight individual sera revealed similar anti-adalimumab antibody concentrations as in the absence of adalimumab, with a Pearson r correlation of 0.99 and an interclass correlation of 0.99. The assay allows accurate evaluation of adalimumab immunogenicity during induction or upon dose intensification and in serum samples not taken at trough.
Collapse
Affiliation(s)
- Sumin Bian
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Campus Gasthuisberg O&N2, PB 820; Herestraat 49, 3000, Leuven, Belgium
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Ann Gils
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Campus Gasthuisberg O&N2, PB 820; Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
46
|
Wendling D, Verhoeven F, Guillot X, Prati C. Immunogenicity of TNF alpha inhibitors in rheumatology: many questions, enough answers? Expert Opin Drug Saf 2016; 16:1-3. [DOI: 10.1080/14740338.2017.1248401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Daniel Wendling
- Department of Rheumatology, University Teaching Hospital, CHRU de Besançon, Besançon, France
- EA4266 and EA4267, Université de Bourgogne Franche-Comté, Besançon, France
| | - Frank Verhoeven
- Department of Rheumatology, University Teaching Hospital, CHRU de Besançon, Besançon, France
- EA4266 and EA4267, Université de Bourgogne Franche-Comté, Besançon, France
| | - Xavier Guillot
- Department of Rheumatology, University Teaching Hospital, CHRU de Besançon, Besançon, France
- EA4266 and EA4267, Université de Bourgogne Franche-Comté, Besançon, France
| | - Clément Prati
- Department of Rheumatology, University Teaching Hospital, CHRU de Besançon, Besançon, France
- EA4266 and EA4267, Université de Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
47
|
Lázár-Molnár E, Delgado JC. Immunogenicity Assessment of Tumor Necrosis Factor Antagonists in the Clinical Laboratory. Clin Chem 2016; 62:1186-98. [DOI: 10.1373/clinchem.2015.242875] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/23/2016] [Indexed: 12/22/2022]
Abstract
Abstract
BACKGROUND
Tumor necrosis factor (TNF) antagonists are increasingly used for the treatment of inflammatory and autoimmune diseases. Immunogenicity of these drugs poses therapeutic challenges such as therapeutic failure and adverse effects in a number of patients. Evaluation of clinical nonresponsiveness includes laboratory testing for drug concentrations and detecting the presence of antidrug antibodies.
CONTENT
This review provides an overview of the immunogenicity of TNF antagonists and testing methodologies currently available for measuring antidrug antibody response, which decreases treatment efficacy and may result in therapeutic failure. This review summarizes methodologies such as binding assays, including ELISA and HPLC-based homogenous mobility shift assay, as well as functional cell-based assays such as reporter gene assay. Furthermore, based on the laboratory findings of testing for antidrug antibody response, as well as serum drug concentrations, an algorithm is provided for interpretation, based on the current available literature and guidelines, which may aid in determining optimal therapy after treatment failure.
SUMMARY
Laboratory testing methodologies for measuring serum concentrations of TNF inhibitors and antidrug antibodies are clinically available. These methods provide an evidence-based, personalized approach for the workup of patients showing treatment failure, which saves time and resources, and contributes to improved patient care.
Collapse
Affiliation(s)
- Eszter Lázár-Molnár
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Julio C Delgado
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
48
|
Schuette A, Moghaddam A, Seemann P, Duda GN, Schmidmaier G, Schomburg L. Treatment with recombinant human bone morphogenetic protein 7 leads to a transient induction of neutralizing autoantibodies in a subset of patients. BBA CLINICAL 2016; 6:100-7. [PMID: 27617228 PMCID: PMC5007422 DOI: 10.1016/j.bbacli.2016.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 01/15/2023]
Abstract
Background Recombinant human bone morphogenetic protein 7 (rhBMP7) is applied for treatment of bone fractures, especially tibial non-unions. Its application may induce autoantibodies (aAB) affecting the targeted and endogenous signaling pathways and in turn negatively impact treatment efficacy. Methods Novel and sensitive assays for the quantification of BMP7-aAB and BMP2-aAB were established and used to analyze serum samples from healthy controls (n = 100 men, n = 100 women) and patients with long bone fracture (n = 265) treated or not with rhBMP7. Sera from three to nine time points per patient were available and enabled the evaluation of aAB over a time course of up to one year. Functional activity of the BMP-aAB was tested with a BMP-responsive cell-based reporter assay. Consolidation of the fracture was evaluated as clinical outcome potentially affected by BMP7-aAB. Results Prevalence of BMP7-aAB and BMP2-aAB was 1–2.5% in non-treated patients or healthy controls. The rhBMP7 treatment induced a transient increase in BMP7-aAB in a subset of patients, returning to non-detectable levels within six months. IgG from BMP7-aAB positive sera inhibited dose dependently the BMP7-reporter gene activity, whereas control sera were without effect. Successful consolidation of the fracture was observed in the majority of both aAB-positive and aAB-negative patients. General significance We conclude that BMP7-aAB can be detected as natural aAB in healthy subjects, and are transiently induced by rhBMP7 therapy in a subset of patients. The aAB are capable of antagonizing BMP7 signaling in vitro, but do not preclude treatment success in patients. There are patients with natural autoantibodies recognizing BMP7. In some patients, rhBMP7-therapy induces BMP7 autoantibodies. BMP7 autoantibodies elicit neutralizing effects on BMP signaling. Therapy-induced BMP7 autoantibodies disappear over time. BMP7 autoantibodies seem not to affect therapy success.
Collapse
Affiliation(s)
- Andrea Schuette
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, D-13353 Berlin, Germany
| | - Arash Moghaddam
- Department of Orthopedics and Trauma Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany
| | - Petra Seemann
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, D-13353 Berlin, Germany
| | - Georg N. Duda
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, D-13353 Berlin, Germany
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, D-13353 Berlin, Germany
| | - Gerhard Schmidmaier
- Department of Orthopedics and Trauma Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, D-13353 Berlin, Germany
- Corresponding author at: Institut für Experimentelle Endokrinologie, Charité - Universitätsmedizin Berlin, Südring 10, CVK, D-13353 Berlin, Germany.Institut für Experimentelle EndokrinologieCharité - Universitätsmedizin BerlinSüdring 10, CVKBerlinD-13353Germany
| |
Collapse
|
49
|
Detection of adalimumab and anti-adalimumab antibodies in patients with rheumatoid arthritis: a comprehensive overview of methodology pitfalls and benefits. Immunol Res 2016; 65:172-185. [DOI: 10.1007/s12026-016-8824-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
50
|
Pelletier AL, Nicaise-Roland P. Adalimumab and pharmacokinetics: Impact on the clinical prescription for inflammatory bowel disease. World J Pharmacol 2016; 5:44-50. [DOI: 10.5497/wjp.v5.i1.44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/19/2015] [Accepted: 12/14/2015] [Indexed: 02/06/2023] Open
Abstract
Anti-tumor necrosis factor (TNF) drugs are widely prescribed for inflammatory disease. A loss of response to adalimumab is frequent and the pharmacokinetics of anti-TNF therapy have important implications for patient management. Individual factors such as albumin, body weight, and disease severity based on the C-reactive protein level influence drug metabolism. Adalimumab trough levels are associated with clinical remission. On the other hand, the detection of antibodies is associated with clinical relapse. Immunosuppressive therapy could reduce antibody formation although the clinical impact is not proven. New algorithms are available to provide personalized treatment and adapt the dosage. More data are needed on dose de-escalation.
Collapse
|