1
|
van Vugt LK, Zwart TC, Bezstarosti S, Heidt S, Reinders MEJ, Hesselink DA, de Vries APJ, de Winter BCM, Moes DJAR. Alemtuzumab Exposure and T Lymphocyte Depletion: A Population Pharmacokinetic-Pharmacodynamic Model of Alemtuzumab Induction Therapy for Kidney Transplantation. Clin Pharmacol Ther 2025. [PMID: 40374867 DOI: 10.1002/cpt.3714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025]
Abstract
Alemtuzumab is a T cell-depleting monoclonal antibody that is used for the prevention of kidney transplant rejection. The duration of lymphodepletion after the current standard induction therapy dose is likely longer than necessary, resulting in prolonged T cell lymphopenia with the associated risk of infections. Here, the interplay between alemtuzumab exposure and T cell dynamics was quantitatively evaluated, and the influence of different doses on T cell recovery was investigated. A population pharmacokinetic-pharmacodynamic model describing the interplay between 30 mg alemtuzumab induction therapy and T cell dynamics in kidney transplantation was developed using NONMEM, using pharmacodynamic data from the Triton study (NCT02057965). The developed model was used to perform an exposure-response analysis and investigate dose optimization with model-derived simulations. In total, 418 peripheral blood T cell measurements from 61 adult kidney transplant recipients were included for model development. A single-compartment turnover Emax model with a first-order T cell influx with feedback and a first-order T cell efflux with parallel alemtuzumab-stimulated T cell removal best described the data. Higher alemtuzumab exposure was associated with lower individual-predicted T cells 4 weeks after administration and longer T cell recovery (> 200 cells/μL). In the simulations, a fixed dose of 15 mg improved median recovery times by 19 days as compared to the standard 30 mg dose without influencing early T cell depletion. A population pharmacokinetic-pharmacodynamic model adequately described T cell dynamics after alemtuzumab induction therapy in kidney transplant recipients. This model can be used to inform future dose-optimization studies of alemtuzumab in different clinical settings.
Collapse
Affiliation(s)
- Lukas K van Vugt
- Erasmus MC Transplant Institute, Rotterdam, The Netherlands
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tom C Zwart
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Hospital Pharmacy, Haga Teaching Hospital, The Hague, The Netherlands
| | - Suzanne Bezstarosti
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sebastiaan Heidt
- Erasmus MC Transplant Institute, Rotterdam, The Netherlands
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marlies E J Reinders
- Erasmus MC Transplant Institute, Rotterdam, The Netherlands
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis A Hesselink
- Erasmus MC Transplant Institute, Rotterdam, The Netherlands
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aiko P J de Vries
- Leiden Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Brenda C M de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Welzel T, Zapf B, Klotsche J, Satirer Ö, Benseler SM, Kuemmerle-Deschner JB. Optimized Treatment of Interleukin (IL-1)-Mediated Autoinflammatory Diseases: Impact of Disease Activity-Based Treatment Adjustments. J Clin Med 2024; 13:2319. [PMID: 38673592 PMCID: PMC11050771 DOI: 10.3390/jcm13082319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Background: Effective control of disease activity in Interleukin-1 autoinflammatory diseases (IL-1 AID) is crucial to prevent damage. The aim was to longitudinally analyze the impact of protocolized disease activity-based treatment adjustments in a real-life cohort. Methods: A single-center study of consecutive children with IL-1 AID followed between January 2016 and December 2019 was performed. Demographics, phenotypes, genotypes, inflammatory markers, physician (PGA), and patient/parent (PPGA) global assessment were captured. Disease activity and treatment changes were assessed. The impact of distinct parameters on disease activity trajectories was analyzed. Results: A total of 56 children were included, median follow-up was 2.1 years reflecting 361 visits. Familial Mediterranean Fever was the most common IL-1 AID. At the first visit, 68% of the patients had moderate/severe disease activity. Disease activity-based treatment adjustments were required in 28/56 children (50%). At last follow-up, 79% had a well-controlled disease. Both PGA and PPGA decreased significantly over time (p < 0.001; p < 0.017, respectively), however, both differed statistically at last visit (p < 0.001). Only PGA showed a significant estimated mean decrease across all IL-1 AID over time. Conclusions: Disease activity-based treatment adjustments can effectively refine treat-to-target strategies, enable personalized precision health approaches, and improve outcomes in children with IL-1 AID.
Collapse
Affiliation(s)
- Tatjana Welzel
- Pediatric Rheumatology, University Children’s Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Pediatric Research Centre, University Children’s Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Division of Pediatric Rheumatology, Department of Pediatrics, autoinflammatory reference centre Tuebingen, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Beate Zapf
- Division of Pediatric Rheumatology, Department of Pediatrics, autoinflammatory reference centre Tuebingen, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Jens Klotsche
- German Rheumatism Research Centre Berlin, 10117 Berlin, Germany
| | - Özlem Satirer
- Division of Pediatric Rheumatology, Department of Pediatrics, autoinflammatory reference centre Tuebingen, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Susanne M. Benseler
- Pediatric Rheumatology, Department of Paediatrics, Alberta Children’s Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Children’s Health Ireland (CHI), D01 R5P3 Dublin, Ireland
| | - Jasmin B. Kuemmerle-Deschner
- Division of Pediatric Rheumatology, Department of Pediatrics, autoinflammatory reference centre Tuebingen, University Hospital Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
3
|
Berkhout LC, I'Ami MJ, Kruithof S, Vogelzang EH, Hooijberg F, Hart MHL, Bentlage AEH, Thomas D, Vermeire S, Vidarsson G, Ten Brinke A, Nurmohamed MT, Wolbink GJ, Rispens T. Formation and clearance of TNF-TNF inhibitor complexes during TNF inhibitor treatment. Br J Pharmacol 2024; 181:1165-1181. [PMID: 37859583 DOI: 10.1111/bph.16269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/29/2023] [Accepted: 07/31/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND AND PURPOSE Millions of patients with inflammatory diseases are treated with tumour necrosis factor (TNF) inhibitors (TNFi). Individual treatment response varies, in part related to variable drug clearance. The role of TNF-TNFi complexes in clearance of the different TNFi is controversial. Moreover, mechanistic insight into the structural aspects and biological significance of TNF-TNFi complexes is lacking. We hypothesized a role for Fc-mediated clearance of TNF-TNFi immune complexes. Therefore, we investigated circulating TNF-TNFi complexes upon treatment with certolizumab-lacking Fc tails-in comparison with adalimumab, golimumab, infliximab and etanercept. EXPERIMENTAL APPROACH Drug-tolerant ELISAs were developed and used to quantify TNF during adalimumab, golimumab, etanercept, certolizumab and infliximab treatment in patients with inflammatory arthritis or ulcerative colitis for a maximum follow-up of 1 year. Effects on in vitro TNF production and Fc-mediated uptake of TNF-TNFi complexes were investigated for all five TNFi. KEY RESULTS Circulating TNF concentrations were >20-fold higher during certolizumab treatment compared with adalimumab, reaching up to 23.1 ng·ml-1 . Internalization of TNF-TNFi complexes by macrophages depended on Fc valency, with efficient uptake for the full antibody TNFi (three Fc tails), but little or no uptake for etanercept and certolizumab (one and zero Fc tail, respectively). TNF production was not affected by TNFi. Total TNF load did not affect clearance rate of total TNFi. CONCLUSIONS AND IMPLICATIONS Differences in TNFi structure profoundly affect clearance of TNF, while it is unlikely that TNF itself significantly contributes to target-mediated drug disposition of TNFi.
Collapse
Affiliation(s)
- Lea Catharina Berkhout
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Merel Jeanne I'Ami
- Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
| | - Simone Kruithof
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik Hans Vogelzang
- Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
| | - Femke Hooijberg
- Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
| | - Margaretha Hendrika Louise Hart
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Arthur Ebel Herman Bentlage
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands
| | - Debby Thomas
- Department of Pharmaceutical and Pharmacological Sciences, Laboratory for Therapeutic and Diagnostic Antibodies, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Severine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Translational Research in Gastrointestinal Disorders, University Hospitals Leuven, Leuven, Belgium
| | - Gestur Vidarsson
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael Twahier Nurmohamed
- Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center | VU University Medical Center, Amsterdam, The Netherlands
| | - Gerrit Jan Wolbink
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center | Reade, Amsterdam, The Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Welzel T, Golhen K, Atkinson A, Gotta V, Ternant D, Kuemmerle-Deschner JB, Michler C, Koch G, van den Anker JN, Pfister M, Woerner A. Prospective study to characterize adalimumab exposure in pediatric patients with rheumatic diseases. Pediatr Rheumatol Online J 2024; 22:5. [PMID: 38167019 PMCID: PMC10763375 DOI: 10.1186/s12969-023-00930-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND In pediatric rheumatic diseases (PRD), adalimumab is dosed using fixed weight-based bands irrespective of methotrexate co-treatment, disease activity (DA) or other factors that might influence adalimumab pharmacokinetics (PK). In rheumatoid arthritis (RA) adalimumab exposure between 2-8 mg/L is associated with clinical response. PRD data on adalimumab is scarce. Therefore, this study aimed to analyze adalimumab PK and its variability in PRD treated with/without methotrexate. METHODS A two-center prospective study in PRD patients aged 2-18 years treated with adalimumab and methotrexate (GA-M) or adalimumab alone (GA) for ≥ 12 weeks was performed. Adalimumab concentrations were collected 1-9 (maximum concentration; Cmax), and 10-14 days (minimum concentration; Cmin) during ≥ 12 weeks following adalimumab start. Concentrations were analyzed with enzyme-linked immunosorbent assay (lower limit of quantification: 0.5 mg/L). Log-normalized Cmin were compared between GA-M and GA using a standard t-test. RESULTS Twenty-eight patients (14 per group), diagnosed with juvenile idiopathic arthritis (71.4%), non-infectious uveitis (25%) or chronic recurrent multifocal osteomyelitis (3.6%) completed the study. GA-M included more females (71.4%; GA 35.7%, p = 0.13). At first study visit, children in GA-M had a slightly longer exposure to adalimumab (17.8 months [IQR 9.6, 21.6]) compared to GA (15.8 months [IQR 8.5, 30.8], p = 0.8). Adalimumab dosing was similar between both groups (median dose 40 mg every 14 days) and observed DA was low. Children in GA-M had a 27% higher median overall exposure compared to GA, although median Cmin adalimumab values were statistically not different (p = 0.3). Cmin values ≥ 8 mg/L (upper limit RA) were more frequently observed in GA-M versus GA (79% versus 64%). Overall, a wide range of Cmin values was observed in PRD (0.5 to 26 mg/L). CONCLUSION This study revealed a high heterogeneity in adalimumab exposure in PRD. Adalimumab exposure tended to be higher with methotrexate co-treatment compared to adalimumab monotherapy although differences were not statistically significant. Most children showed adalimumab exposure exceeding those reported for RA with clinical response, particularly with methotrexate co-treatment. This highlights the need of further investigations to establish model-based personalized treatment strategies in PRD to avoid under- and overexposure. TRIAL REGISTRATION NCT04042792 , registered 02.08.2019.
Collapse
Affiliation(s)
- Tatjana Welzel
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland.
- Division of Pediatric Rheumatology, Department of Pediatrics and Autoinflammation Reference Centre Tuebingen (arcT), University Hospital Tuebingen, Tuebingen, Germany.
- Pediatric Rheumatology, University Children's Hospital Basel, University of Basel, Basel, Switzerland.
- Pediatric Research Center, University Children's Hospital Basel, University of Basel, Basel, Switzerland.
| | - Klervi Golhen
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrew Atkinson
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Pediatric Research Center, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Division of Infectious Diseases, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Verena Gotta
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - David Ternant
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Université de Tours, service de pharmacologie médicale, Tours France, Université de Tours, EA 4245 T2I, Tours, France
| | - Jasmin B Kuemmerle-Deschner
- Division of Pediatric Rheumatology, Department of Pediatrics and Autoinflammation Reference Centre Tuebingen (arcT), University Hospital Tuebingen, Tuebingen, Germany
| | - Christine Michler
- Division of Pediatric Rheumatology, Department of Pediatrics and Autoinflammation Reference Centre Tuebingen (arcT), University Hospital Tuebingen, Tuebingen, Germany
| | - Gilbert Koch
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Johannes N van den Anker
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Andreas Woerner
- Pediatric Rheumatology, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
5
|
Lobet S, Paintaud G, Azzopardi N, Passot C, Caulet M, Chautard R, Desvignes C, Capitain O, Tougeron D, Lecomte T, Ternant D. Relationship Between Cetuximab Target-Mediated Pharmacokinetics and Progression-Free Survival in Metastatic Colorectal Cancer Patients. Clin Pharmacokinet 2023; 62:1263-1274. [PMID: 37442917 DOI: 10.1007/s40262-023-01270-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Cetuximab, an anti-epidermal growth factor receptor (EGFR) monoclonal immunoglobulin (Ig)G1 antibody, has been approved for the treatment of metastatic colorectal cancer (mCRC). The influence of target-antigen on cetuximab pharmacokinetics has never been investigated using target-mediated drug disposition (TMDD) modelling. This study aimed to investigate the relationship between cetuximab concentrations, target kinetics and progression-free survival (PFS). METHODS In this ancillary study (NCT00559741), 91 patients with mCRC treated with cetuximab were assessed. Influence of target levels on cetuximab pharmacokinetics was described using TMDD modelling. The relationship between cetuximab concentrations, target kinetics and time-to-progression (TTP) was described using a joint pharmacokinetic-TTP model, where unbound target levels were assumed to influence hazard of progression by an Emax model. Mitigation strategies of concentration-response relationship, i.e., time-varying endogenous clearance and mutual influences of clearance and time-to-progression were investigated. RESULTS Cetuximab concentration-time data were satisfactorily described using the TMDD model with quasi-steady-state approximation and time-varying endogenous clearance. Estimated target parameters were baseline target levels (R0 = 43 nM), and complex elimination rate constant (kint = 0.95 day-1). Estimated time-varying clearance parameters were time-invariant component of CL (CL0= 0.38 L/day-1), time-variant component of CL (CL1= 0.058 L/day-1) and first-order rate of CL1 decreasing over time (kdes = 0.049 day-1). Part of concentration-TTP was TTP-driven, where clearance and TTP were inversely correlated. In addition, increased target occupancy was associated with increased TTP. CONCLUSION This is the first study describing the complex relationship between cetuximab target-mediated pharmacokinetics and PFS in mCRC patients using a joint PK-time-to-progression model. Further studies are needed to provide a more in-depth description of this relationship.
Collapse
Affiliation(s)
- Sarah Lobet
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Tours University, Tours, France
| | - Gilles Paintaud
- EA4245 Transplantation, Immunologie, Inflammation (T2i), Tours University, Tours, France
- Centre Pilote de suivi Biologique des traitements par Anticorps (CePiBAc), Tours University Hospital, Tours, France
- Pharmacology-Toxicology Department, Tours University Hospital, Tours, France
| | | | - Christophe Passot
- Oncopharmacology-Pharmacogenetics Department INSERM U892, Institut de Cancérologie de l'Ouest site Paul Papin, Angers, France
| | - Morgane Caulet
- Gastroenterology and Digestive oncology Department, Tours University Hospital, Tours, France
| | - Romain Chautard
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Tours University, Tours, France
- Gastroenterology and Digestive oncology Department, Tours University Hospital, Tours, France
| | - Céline Desvignes
- EA4245 Transplantation, Immunologie, Inflammation (T2i), Tours University, Tours, France
- Centre Pilote de suivi Biologique des traitements par Anticorps (CePiBAc), Tours University Hospital, Tours, France
| | - Olivier Capitain
- Oncopharmacology-Pharmacogenetics Department INSERM U892, Institut de Cancérologie de l'Ouest site Paul Papin, Angers, France
| | - David Tougeron
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France
- PRoDiCeT, Poitiers University, Poitiers, France
| | - Thierry Lecomte
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Tours University, Tours, France
- Gastroenterology and Digestive oncology Department, Tours University Hospital, Tours, France
| | - David Ternant
- EA4245 Transplantation, Immunologie, Inflammation (T2i), Tours University, Tours, France.
- Centre Pilote de suivi Biologique des traitements par Anticorps (CePiBAc), Tours University Hospital, Tours, France.
- Pharmacology-Toxicology Department, Tours University Hospital, Tours, France.
| |
Collapse
|
6
|
Busto-Iglesias M, Rodríguez-Martínez L, Rodríguez-Fernández CA, González-López J, González-Barcia M, de Domingo B, Rodríguez-Rodríguez L, Fernández-Ferreiro A, Mondelo-García C. Perspectives of Therapeutic Drug Monitoring of Biological Agents in Non-Infectious Uveitis Treatment: A Review. Pharmaceutics 2023; 15:pharmaceutics15030766. [PMID: 36986627 PMCID: PMC10051556 DOI: 10.3390/pharmaceutics15030766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Biological drugs, especially those targeting anti-tumour necrosis factor α (TNFα) molecule, have revolutionized the treatment of patients with non-infectious uveitis (NIU), a sight-threatening condition characterized by ocular inflammation that can lead to severe vision threatening and blindness. Adalimumab (ADA) and infliximab (IFX), the most widely used anti-TNFα drugs, have led to greater clinical benefits, but a significant fraction of patients with NIU do not respond to these drugs. The therapeutic outcome is closely related to systemic drug levels, which are influenced by several factors such as immunogenicity, concomitant treatment with immunomodulators, and genetic factors. Therapeutic drug monitoring (TDM) of drug and anti-drug antibody (ADAbs) levels is emerging as a resource to optimise biologic therapy by personalising treatment to bring and maintain drug concentration within the therapeutic range, especially in those patients where a clinical response is less than expected. Furthermore, some studies have described different genetic polymorphisms that may act as predictors of response to treatment with anti-TNFα agents in immune-mediated diseases and could be useful in personalising biologic treatment selection. This review is a compilation of the published evidence in NIU and in other immune-mediated diseases that support the usefulness of TDM and pharmacogenetics as a tool to guide clinicians’ treatment decisions leading to better clinical outcomes. In addition, findings from preclinical and clinical studies, assessing the safety and efficacy of intravitreal administration of anti-TNFα agents in NIU are discussed.
Collapse
Affiliation(s)
- Manuel Busto-Iglesias
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain (C.M.-G.)
- Pharmacology Group, Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| | - Lorena Rodríguez-Martínez
- Pharmacology Group, Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| | - Carmen Antía Rodríguez-Fernández
- Pharmacology Group, Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
- Ophthalmology Department, Bellvitge University Hospital, 08907 Barcelona, Spain
| | - Jaime González-López
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain (C.M.-G.)
- Pharmacology Group, Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| | - Miguel González-Barcia
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain (C.M.-G.)
- Pharmacology Group, Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| | - Begoña de Domingo
- Ophthalmology Department, University Clinical Hospital of Santiago Compostela (SERGAS), 15706 Santiago de Compostela, Spain
| | - Luis Rodríguez-Rodríguez
- Musculoskeletal Pathology Group, Hospital Clínico San Carlos, Instituto Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
- Correspondence: (L.R.-R.); (A.F.-F.)
| | - Anxo Fernández-Ferreiro
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain (C.M.-G.)
- Pharmacology Group, Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
- Correspondence: (L.R.-R.); (A.F.-F.)
| | - Cristina Mondelo-García
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain (C.M.-G.)
- Pharmacology Group, Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
7
|
Welzel T, Oefelein L, Twilt M, Pfister M, Kuemmerle-Deschner JB, Benseler SM. Tapering of biological treatment in autoinflammatory diseases: a scoping review. Pediatr Rheumatol Online J 2022; 20:67. [PMID: 35964053 PMCID: PMC9375310 DOI: 10.1186/s12969-022-00725-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Biological treatment and treat-to-target approaches guide the achievement of inactive disease and clinical remission in Autoinflammatory Diseases (AID). However, there is limited evidence addressing optimal tapering strategies and/or discontinuation of biological treatment in AID. This study evaluates available evidence of tapering biological treatment and explores key factors for successful tapering. METHODS A systematic literature search was conducted in Embase, MEDLINE, Cochrane Database of Systematic Reviews and Cochrane Central Register of Controlled Trials using the OVID platform (1990-08/2020). Bibliographic search of relevant reviews was also performed. Studies/case series (n ≥ 5) in AID patients aged ≤ 18 years with biological treatment providing information on tapering/treatment discontinuation were included. After quality assessment aggregated data were extracted and synthesized. Tapering strategies were explored. RESULTS A total of 6035 records were identified. Four papers were deemed high quality, all focused on systemic juvenile idiopathic arthritis (sJIA) (1 open-label randomized trial, 2 prospective, 1 retrospective observational study). Biological treatment included anakinra (n = 2), canakinumab (n = 1) and tocilizumab (n = 1). Strategies in anakinra tapering included alternate-day regimen. Canakinumab tapering was performed randomized for dose reduction or interval prolongation, whereas tocilizumab was tapered by interval prolongation. Key factors identified included early start of biological treatment and sustained inactive disease. CONCLUSION Tapering of biological treatment after sustained inactive disease should be considered. Guidance for optimal strategies is limited. Future studies may leverage therapeutic drug monitoring in combination with pharmacometric modelling to further enhance personalized "taper-to-target" strategies respecting individual patients and diseases aspects.
Collapse
Affiliation(s)
- Tatjana Welzel
- Pediatric Rheumatology and autoinflammation reference center Tuebingen (arcT), Department of Pediatrics, Member of the European Reference Network for rare or low prevalence complex diseases, network Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN RITA), University Children`s Hospital Tuebingen, Tuebingen, Germany.
- Pediatric Pharmacology and Pharmacometrics, Pediatric Rheumatology, University Children`s Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, CH, 4031, Basel, Switzerland.
| | - Lea Oefelein
- Pediatric Rheumatology and autoinflammation reference center Tuebingen (arcT), Department of Pediatrics, Member of the European Reference Network for rare or low prevalence complex diseases, network Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN RITA), University Children`s Hospital Tuebingen, Tuebingen, Germany
| | - Marinka Twilt
- Rheumatology, Department of Pediatrics, Alberta Children`s Hospital, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, Pediatric Rheumatology, University Children`s Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, CH, 4031, Basel, Switzerland
| | - Jasmin B Kuemmerle-Deschner
- Pediatric Rheumatology and autoinflammation reference center Tuebingen (arcT), Department of Pediatrics, Member of the European Reference Network for rare or low prevalence complex diseases, network Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN RITA), University Children`s Hospital Tuebingen, Tuebingen, Germany
| | - Susanne M Benseler
- Rheumatology, Department of Pediatrics, Alberta Children`s Hospital, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
8
|
Le Tilly O, Gatault P, Baron C, Bejan‐Angoulvant T, Büchler M, Paintaud G, Ternant D. Is CD25 blockade optimal in kidney transplant patients treated with basiliximab? A target‐mediated drug disposition (TMDD) model. Br J Clin Pharmacol 2022; 88:3500-3505. [DOI: 10.1111/bcp.15235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/02/2022] [Accepted: 01/09/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Olivier Le Tilly
- Université de Tours, EA 4245 Transplantation, Immunology, Inflammation Tours France
- CHRU de Tours Service de Pharmacologie Médicale Tours France
| | - Philippe Gatault
- Université de Tours, EA 4245 Transplantation, Immunology, Inflammation Tours France
- CHRU de Tours, Department of Nephrology and Transplantation Tours France
| | - Christophe Baron
- Université de Tours, EA 4245 Transplantation, Immunology, Inflammation Tours France
- CHRU de Tours, Department of Nephrology and Transplantation Tours France
| | - Theodora Bejan‐Angoulvant
- Université de Tours, EA 4245 Transplantation, Immunology, Inflammation Tours France
- CHRU de Tours Service de Pharmacologie Médicale Tours France
| | - Matthias Büchler
- Université de Tours, EA 4245 Transplantation, Immunology, Inflammation Tours France
- CHRU de Tours, Department of Nephrology and Transplantation Tours France
| | - Gilles Paintaud
- Université de Tours, EA 4245 Transplantation, Immunology, Inflammation Tours France
- CHRU de Tours Service de Pharmacologie Médicale Tours France
| | - David Ternant
- Université de Tours, EA 4245 Transplantation, Immunology, Inflammation Tours France
- CHRU de Tours Service de Pharmacologie Médicale Tours France
| |
Collapse
|
9
|
Peckys DB, Gaa D, de Jonge N. Quantification of EGFR-HER2 Heterodimers in HER2-Overexpressing Breast Cancer Cells Using Liquid-Phase Electron Microscopy. Cells 2021; 10:cells10113244. [PMID: 34831465 PMCID: PMC8623301 DOI: 10.3390/cells10113244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Currently, breast cancer patients are classified uniquely according to the expression level of hormone receptors, and human epidermal growth factor receptor 2 (HER2). This coarse classification is insufficient to capture the phenotypic complexity and heterogeneity of the disease. A methodology was developed for absolute quantification of receptor surface density ρR, and molecular interaction (dimerization), as well as the associated heterogeneities, of HER2 and its family member, the epidermal growth factor receptor (EGFR) in the plasma membrane of HER2 overexpressing breast cancer cells. Quantitative, correlative light microscopy (LM) and liquid-phase electron microscopy (LPEM) were combined with quantum dot (QD) labeling. Single-molecule position data of receptors were obtained from scanning transmission electron microscopy (STEM) images of intact cancer cells. Over 280,000 receptor positions were detected and statistically analyzed. An important finding was the subcellular heterogeneity in heterodimer shares with respect to plasma membrane regions with different dynamic properties. Deriving quantitative information about EGFR and HER2 ρR, as well as their dimer percentages, and the heterogeneities thereof, in single cancer cells, is potentially relevant for early identification of patients with HER2 overexpressing tumors comprising an enhanced share of EGFR dimers, likely increasing the risk for drug resistance, and thus requiring additional targeted therapeutic strategies.
Collapse
Affiliation(s)
- Diana B. Peckys
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, 66421 Homburg, Germany;
| | - Daniel Gaa
- INM—Leibniz Institute for New Materials, 66123 Saarbrücken, Germany;
| | - Niels de Jonge
- INM—Leibniz Institute for New Materials, 66123 Saarbrücken, Germany;
- Department of Physics, Saarland University, 66123 Saarbrücken, Germany
- Correspondence:
| |
Collapse
|
10
|
Bensalem A, Cartron G, Specks U, Mulleman D, Gyan E, Cornec D, Desvignes C, Casasnovas O, Lamy T, Leprêtre S, Paintaud G, Ternant D. The Influence of Underlying Disease on Rituximab Pharmacokinetics May be Explained by Target-Mediated Drug Disposition. Clin Pharmacokinet 2021; 61:423-437. [PMID: 34773607 DOI: 10.1007/s40262-021-01081-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVES Rituximab is an anti-CD20 monoclonal antibody approved in several diseases, including chronic lymphocytic leukemia (CLL), diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), rheumatoid arthritis (RA), and anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV). The influence of underlying disease on rituximab pharmacokinetics has never been investigated for several cancer and non-cancer diseases simultaneously. This study aimed at assessing this influence using an integrated semi-mechanistic model accounting for target-mediated elimination of rituximab. METHODS Rituximab concentration-time data from five studies previously published in patients with CLL, DLBCL, FL, RA, and AAV were described using a two-compartment model with irreversible binding of rituximab to its target antigen. Both underlying disease and target antigen measurements were assessed as covariates. RESULTS Central volume of distribution was [95% confidence interval] 1.7-fold [1.6-1.9] higher in DLBCL than in RA, FL, and CLL, and it was 1.8-fold [1.6-2.1] higher in RA, FL, and CLL than in AAV. First-order elimination rate constants were 1.8-fold [1.7-2.0] and 1.3-fold [1.2-1.5] higher in RA, DLBCL, and FL than in CLL and AAV, respectively. Baseline latent antigen level (L0) was 54-fold [30-94], 20-fold [11-36], and 29-fold [14-64] higher in CLL, DLBCL, and FL, respectively, than in RA and AAV. In lymphoma, L0 increased with baseline total metabolic tumor volume (p = 6.10-7). In CLL, the second-order target-mediated elimination rate constant (kdeg) increased with baseline CD20 count on circulating B cells (CD20cir, p = 0.0081). CONCLUSIONS Our results show for the first time that rituximab pharmacokinetics is strongly influenced by underlying disease and disease activity. Notably, neoplasms are associated with higher antigen amounts that result in decreased exposure to rituximab compared to inflammatory diseases. Our model might be used to estimate unbound target amounts in upcoming studies.
Collapse
MESH Headings
- Antigens, CD20/metabolism
- Arthritis, Rheumatoid/drug therapy
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Lymphoma, Follicular/drug therapy
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Rituximab/pharmacokinetics
Collapse
Affiliation(s)
| | - Guillaume Cartron
- CNRS UMR 5235, Université de Montpellier, Montpellier, France
- Department of Hematology, CHRU Montpellier, Montpellier, France
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Denis Mulleman
- Université de Tours, EA 7501 GICC, Tours, France
- Department of Rheumatology, CHRU de Tours, Tours, France
| | - Emmanuel Gyan
- Department of Hematology and Cell Therapy, Clinical Investigations Center INSERM U1415, CHU Tours, Tours, France
| | - Divi Cornec
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
- Rheumatology Department, Brest University Hospital, and INSERM U1227, Brest, France
| | - Celine Desvignes
- Université de Tours, EA 4245 T2I, Tours, France
- Service de Pharmacologie Médicale, CHU Bretonneau, 2 Boulevard Tonnellé, 37044, Tours, France
| | - Olivier Casasnovas
- Department of Clinical Hematology, CHU Dijon, Dijon, France
- INSERM Lipids, Nutrition, Cancer (LNC) UMR 866, Dijon, France
| | - Thierry Lamy
- Department of Clinical Hematology, CHU Rennes, U917, Rennes, France
| | | | - Gilles Paintaud
- Université de Tours, EA 4245 T2I, Tours, France
- Service de Pharmacologie Médicale, CHU Bretonneau, 2 Boulevard Tonnellé, 37044, Tours, France
| | - David Ternant
- Université de Tours, EA 4245 T2I, Tours, France.
- Service de Pharmacologie Médicale, CHU Bretonneau, 2 Boulevard Tonnellé, 37044, Tours, France.
| |
Collapse
|
11
|
Ternant D, Le Tilly O, Picon L, Moussata D, Passot C, Bejan-Angoulvant T, Desvignes C, Mulleman D, Goupille P, Paintaud G. Infliximab Efficacy May Be Linked to Full TNF-α Blockade in Peripheral Compartment-A Double Central-Peripheral Target-Mediated Drug Disposition (TMDD) Model. Pharmaceutics 2021; 13:pharmaceutics13111821. [PMID: 34834236 PMCID: PMC8623740 DOI: 10.3390/pharmaceutics13111821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/03/2022] Open
Abstract
Infliximab is an anti-TNF-α monoclonal antibody approved in chronic inflammatory bowel diseases (IBD). This study aimed at providing an in-depth description of infliximab target-mediated pharmacokinetics in 133 IBD patients treated with 5 mg/kg infliximab at weeks 0, 2, 14, and 22. A two-compartment model with double target-mediated drug disposition (TMDD) in both central and peripheral compartments was developed, using a rich database of 26 ankylosing spondylitis patients as a reference for linear elimination kinetics. Population approach and quasi-steady-state (QSS) approximation were used. Concentration-time data were satisfactorily described using the double-TMDD model. Target-mediated parameters of central and peripheral compartments were respectively baseline TNF concentrations (RC0 = 3.3 nM and RP0 = 0.46 nM), steady-stated dissociation rates (KCSS = 15.4 nM and KPSS = 0.49 nM), and first-order elimination rates of complexes (kCint = 0.17 day−1 and kPint = 0.0079 day−1). This model showed slower turnover of targets and infliximab-TNF complex elimination rate in peripheral compartment than in central compartment. This study allowed a better understanding of the multi-scale target-mediated pharmacokinetics of infliximab. This model could be useful to improve model-based therapeutic drug monitoring of infliximab in IBD patients.
Collapse
Affiliation(s)
- David Ternant
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
- Correspondence:
| | - Olivier Le Tilly
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| | - Laurence Picon
- Department of Gastroenterology, CHRU de Tours, 37044 Tour, France;
| | - Driffa Moussata
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Gastroenterology, CHRU de Tours, 37044 Tour, France;
| | - Christophe Passot
- Département de Biopathologie, Institut de Cancérologie de l’Ouest, 49055 Angers, France;
| | - Theodora Bejan-Angoulvant
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| | - Céline Desvignes
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| | - Denis Mulleman
- EA 7501 “Groupe Innovation et Ciblage Cellulaire”, Université de Tours, 37044 Tour, France;
- Department of Rheumatology, CHRU de Tours, 37044 Tour, France;
| | | | - Gilles Paintaud
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| |
Collapse
|
12
|
Infliximab Treatment Does Not Lead to Full TNF-α Inhibition: A Target-Mediated Drug Disposition Model. Clin Pharmacokinet 2021; 61:143-154. [PMID: 34351609 DOI: 10.1007/s40262-021-01057-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND OBJECTIVE Infliximab, an anti-tumour necrosis factor (TNF)-α monoclonal antibody, has been approved in chronic inflammatory disease, including rheumatoid arthritis, Crohn's disease and ankylosing spondylitis. This study aimed to investigate and characterise target-mediated drug disposition of infliximab and antigen mass turnover during infliximab treatment. METHODS In this retrospective cohort of 186 patients treated with infliximab for rheumatoid arthritis, Crohn's disease or ankylosing spondylitis, trough infliximab concentrations were determined from samples collected between weeks 0 and 22 after treatment initiation. Target-mediated pharmacokinetics of infliximab was described using target-mediated drug disposition modelling. Target-mediated elimination parameters were determined for rheumatoid arthritis and Crohn's disease, assuming ankylosing spondylitis with no target-mediated elimination. RESULTS The quasi-equilibrium approximation of a target-mediated drug disposition model allowed a satisfactory description of infliximab concentration-time data. Estimated baseline TNF-α amounts were similar in Crohn's disease and rheumatoid arthritis (R0 = 0.39 vs 0.46 nM, respectively), but infliximab-TNF complex elimination was slower in Crohn's disease than in rheumatoid arthritis (kint = 0.024 vs 0.061 day-1, respectively). Terminal elimination half-lives were 13.5, 21.5 and 16.5 days for rheumatoid arthritis, Crohn's disease and ankylosing spondylitis, respectively. Estimated amounts of free target were close to baseline values before the next infusion suggesting that TNF-α inhibition may not be sustained over the entire dose interval. CONCLUSIONS The present study is the first to quantify the influence of target antigen dynamics on infliximab pharmacokinetics. Target-mediated elimination of infliximab may be complex, involving a multi-scale turnover of TNF-α, especially in patients with Crohn's disease. Additional clinical studies are warranted to further evaluate and fine-tune dosing approaches to ensure sustained TNF-α inhibition.
Collapse
|
13
|
Peña-Cabia S, Royuela Vicente A, Ramos Díaz R, Gutiérrez Nicolás F, Peñalver Vera Á, Siso García I, Hitt Sabag R, García Lacalle C, Peña-Cabia A, Iglesias-Peinado I, García Díaz B, López-Martín A. Assessment of exposure-response relationship for bevacizumab in patients with metastatic colorectal cancer. Biomed Pharmacother 2021; 141:111827. [PMID: 34153845 DOI: 10.1016/j.biopha.2021.111827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/27/2022] Open
Abstract
Limited literature is available for bevacizumab exposure-response relationship and there is not a concentration threshold associated with an optimal disease control. This prospective observational study in patients with metastatic colorectal cancer (mCRC) aims to evaluate, in a real-life setting, the relationship between bevacizumab through concentrations at steady state (Ctrough, SS) and disease control. Ctrough, SS were drawn, coinciding with the radiological evaluation of the response (progression or clinical benefit). Generalized estimating equations (GEE) analysis was performed. To test the association between Ctrough, SS in each patient with overall survival (OS) or progression-free survival (PFS), Cox proportional hazard models were developed. Data included 50 bevacizumab Ctrough, SS from 27 patients. The GEE model did not suggest any positive association between bevacizumab Ctrough, SS and clinical benefit (OR 0.99, 95% CI: 0.98-1.02, p = 0.863). The Cox regression showed association between higher median Ctrough, SS with better OS (HR 0.86, 95% CI: 0.73-1.01, p = 0.060), but not with PFS. We cannot confirm a relationship between bevacizumab Ctrough, SS and clinical benefit but this is the first real-world study trying to show a relationship between bevacizumab Ctrough, SS and disease control in mCRC. It was conducted in a small sample size which reduces the level of evidence. Further controlled randomized studies with a sufficient number of patients are required.
Collapse
Affiliation(s)
| | - Ana Royuela Vicente
- Biostatistics Unit, Puerta de Hierro Biomedical Research Institute (IDIPHISA), CIBERESP, Madrid, Spain
| | - Ruth Ramos Díaz
- Foundation Health Research Institute of Canary (FIISC), University Hospital Complex of Canary (CHUC), Tenerife, Spain
| | | | | | | | | | | | - Ana Peña-Cabia
- Medical Laboratory Unit, Virgen de la Luz Hospital, Cuenca, Spain
| | - Irene Iglesias-Peinado
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | | | - Ana López-Martín
- Medical Oncology Unit, Severo Ochoa University Hospital, Madrid, Spain
| |
Collapse
|
14
|
Malik PRV, Temrikar ZH, Chelle P, Edginton AN, Meibohm B. Pediatric Dose Selection for Therapeutic Proteins. J Clin Pharmacol 2021; 61 Suppl 1:S193-S206. [PMID: 34185910 DOI: 10.1002/jcph.1829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022]
Abstract
In selecting optimal dosing regimens in support of the clinical use of monoclonal antibodies and other therapeutic proteins in pediatric indications, the unique pharmacokinetic properties of this class of biologics, as well as the underlying physiologic and pathophysiologic processes and their modulation by childhood growth and development, needs to be appreciated. During drug development, first-in-pediatric dose selection is a capstone event in the pediatric investigation plan that relies heavily on extrapolation of pharmacokinetic and pharmacodynamic data from adult to pediatric populations. It is facilitated by combinations of pharmacometric approaches, including allometry, physiologically based pharmacokinetic modeling, and population pharmacokinetic analyses, although data on reliability and qualification of some of these tools in the context of therapeutic proteins are still limited but emerging. Presented data suggest nonlinear relationships between body weight and both clearance and volume of distribution for therapeutic proteins in pediatric populations, with allometric exponents of 0.75 and 0.8, respectively. For newborns and infants (<1 year), even higher nonlinearity seems to occur. Translation of the quantitative characterization of the pediatric pharmacokinetics of therapeutic proteins into dosing regimens for the drug label requires compromising between precision dosing and clinical practicability, with tiered dosing algorithms based on size or age strata being the currently most frequently applied methodology.
Collapse
Affiliation(s)
- Paul R V Malik
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Zaid H Temrikar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Pierre Chelle
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
15
|
Bensalem A, Mulleman D, Paintaud G, Azzopardi N, Gouilleux-Gruart V, Cornec D, Specks U, Ternant D. Non-Linear Rituximab Pharmacokinetics and Complex Relationship between Rituximab Concentrations and Anti-Neutrophil Cytoplasmic Antibodies (ANCA) in ANCA-Associated Vasculitis: The RAVE Trial Revisited. Clin Pharmacokinet 2021; 59:519-530. [PMID: 31586310 DOI: 10.1007/s40262-019-00826-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND OBJECTIVES Rituximab is approved in patients with anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) and leads to a decrease of ANCA levels. The objectives of this study were to investigate the non-linear pharmacokinetics of rituximab and the relationship between its concentrations and ANCA levels in AAV patients. METHODS Ninety-two AAV patients from the RAVE (Rituximab in ANCA-Associated Vasculitis) trial were assessed. Both ANCA anti-myeloperoxidase (MPO-ANCA) and anti-proteinase 3 (PR3-ANCA) levels were used as biomarkers. The pharmacokinetics of rituximab were described using a semi-mechanistic two-compartment model that included a latent target antigen turnover and allowed the estimation of specific target-mediated elimination in addition to its non-specific elimination of rituximab. The effect of rituximab on the ANCA level was described using a semi-mechanistic compartment model with a negative feedback (Friberg) model with no transit compartment. A population modeling approach was used. RESULTS Our pharmacokinetic and pharmacokinetic-pharmacodynamic (PK-PD) models satisfactorily described both concentration-time and concentration-effect relationship data. The mean (inter-individual standard deviation) estimated non-specific clearance was 0.15 L/day (0.30%) and the target-mediated elimination rate constant was 2.4 × 10-5 nmol/day. The elimination half-lives for MPO-ANCA and PR3-ANCA were 24 and 18 days, respectively. CONCLUSIONS A non-linear target-mediated elimination of rituximab was detected in AAV patients. Our PK-PD model allowed quantification of the association between rituximab concentrations and ANCA levels. This decrease was deep but delayed, and more sustained in patients with MPO-ANCA than in those with PR3-ANCA. Our results suggest that repeating courses of rituximab might improve the clinical response to rituximab.
Collapse
Affiliation(s)
| | - Denis Mulleman
- Université de Tours, EA 7501 GICC, Tours, France.,Department of Rheumatology, CHRU de Tours, Tours, France
| | - Gilles Paintaud
- Université de Tours, EA 7501 GICC, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours, France
| | - Nicolas Azzopardi
- Université de Tours, EA 7501 GICC, Tours, France.,CNRS, ERL 7001, Tours, France
| | - Valérie Gouilleux-Gruart
- Université de Tours, EA 7501 GICC, Tours, France.,Laboratory of Immunology, CHRU de Tours, Tours, France
| | - Divi Cornec
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA.,Rheumatology Department, Brest University Hospital, Brest, France.,INSERM U1227, Brest, France
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - David Ternant
- Université de Tours, EA 7501 GICC, Tours, France. .,Department of Medical Pharmacology, CHRU de Tours, Tours, France. .,Laboratoire de Pharmacologie-toxicologie, CHRU de Tours, 2 Boulevard Tonnellé, 37044, Tours Cedex, France.
| |
Collapse
|
16
|
Therapeutic Antibodies for the Treatment of Respiratory Tract Infections-Current Overview and Perspectives. Vaccines (Basel) 2021; 9:vaccines9020151. [PMID: 33668613 PMCID: PMC7917879 DOI: 10.3390/vaccines9020151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Respiratorytract infections (RTIs) are frequent and life-threatening diseases, accounting for several millions of deaths worldwide. RTIs implicate microorganisms, including viruses (influenza virus, coronavirus, respiratory syncytial virus (RSV)), bacteria (Pseudomonas aeruginosa, Streptococcus pneumoniae, Staphylococcus aureus and Bacillus anthracis) and fungi (Pneumocystis spp., Aspergillus spp. and very occasionally Candida spp.). The emergence of new pathogens, like the coronavirus SARS-CoV-2, and the substantial increase in drug resistance have highlighted the critical necessity to develop novel anti-infective molecules. In this context, antibodies (Abs) are becoming increasingly important in respiratory medicine and may fulfill the unmet medical needs of RTIs. However, development of Abs for treating infectious diseases is less advanced than for cancer and inflammatory diseases. Currently, only three Abs have been marketed for RTIs, namely, against pulmonary anthrax and RSV infection, while several clinical and preclinical studies are in progress. This article gives an overview of the advances in the use of Abs for the treatment of RTIs, based on the analysis of clinical studies in this field. It describes the Ab structure, function and pharmacokinetics, and discusses the opportunities offered by the various Ab formats, Ab engineering and co-treatment strategies. Including the most recent literature, it finally highlights the strengths, weaknesses and likely future trends of a novel anti-RTI Ab armamentarium.
Collapse
|
17
|
Passot C, Sberro-Soussan R, Bertrand D, Caillard S, Schvartz B, Domenger C, Contin-Bordes C, Paintaud G, Halimi JM, Ternant D, Gatault P. Feasibility and safety of tailored dosing schedule for eculizumab based on therapeutic drug monitoring: Lessons from a prospective multicentric study. Br J Clin Pharmacol 2020; 87:2236-2246. [PMID: 33118186 DOI: 10.1111/bcp.14627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 12/23/2022] Open
Abstract
AIMS Eculizumab is an anti-C5 monoclonal antibody approved for rare diseases including atypical haemolytic-uraemic syndrome. The maintenance phase dosing regimen is identical for all adult patients: 1200 mg every 2 weeks. Recent studies reported an overexposure in many patients when considering a target trough concentration range of 50-100 mg/L. The aim of the present work was to validate the feasibility of therapeutic drug monitoring of eculizumab in atypical haemolytic-uraemic syndrome patients. METHODS We performed a 2-step prospective multicentre study. In the first phase, we developed a pharmacokinetic population model using data from 40 patients and identified patients for whom a 1-week lengthening of interval between infusions would lead to a trough concentration above 100 mg/L. In the second phase, selected patients were allocated a 1-week extension and eculizumab trough concentrations were monitored. RESULTS The model confirmed the previously reported influence of bodyweight on elimination clearance and predicted that 36 (90%) patients would be eligible for interval extension. In the second phase of the study, a 1-week lengthening of interval between infusions was performed in 15 patients whose trough concentration at the next visit was predicted with a Bayesian model to be above 100 mg/L. After interval extension, 10 patients (67%) presented measured trough concentrations over 100 mg/L. No biological or clinical recurrence of disease was observed, even in the 5 patients with concentrations below 100 mg/L in whom the initial dosing regimen was resumed. CONCLUSION Safe eculizumab interval adjustment is feasible with a PK monitoring.
Collapse
Affiliation(s)
- Christophe Passot
- EA7501, University of Tours, France.,Laboratory of Pharmacology-Toxicology, CHRU de Tours, France.,Integrated Center for Oncology, Angers, France
| | - Rebecca Sberro-Soussan
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Sophie Caillard
- Nephrology and Transplant Department, Strasbourg University Hospital, Strasbourg, France
| | | | | | | | - Gilles Paintaud
- EA7501, University of Tours, France.,Laboratory of Pharmacology-Toxicology, CHRU de Tours, France
| | - Jean-Michel Halimi
- EA4245 Transplant Immunology and Inflammation, Université de Tours, Tours, France.,Service de Néphrologie-Hypertension artérielle, Dialyses, Transplantation rénale, CHRU Tours, France
| | - David Ternant
- EA7501, University of Tours, France.,Laboratory of Pharmacology-Toxicology, CHRU de Tours, France
| | - Philippe Gatault
- EA4245 Transplant Immunology and Inflammation, Université de Tours, Tours, France.,Service de Néphrologie-Hypertension artérielle, Dialyses, Transplantation rénale, CHRU Tours, France
| |
Collapse
|
18
|
Le Tilly O, Bejan-Angoulvant T, Paintaud G, Ternant D. Letter to Dreesen et al. on their article "Modelling of the Relationship Between Infliximab Exposure, Faecal Calprotectin, and Endoscopic Remission in Patients With Crohn's Disease"-A comprehensive review of infliximab population pharmacokinetic modelling publications. Br J Clin Pharmacol 2020; 87:1594-1595. [PMID: 33269471 DOI: 10.1111/bcp.14554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/31/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Olivier Le Tilly
- Université de Tours, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours, France
| | - Theodora Bejan-Angoulvant
- Université de Tours, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours, France
| | - Gilles Paintaud
- Université de Tours, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours, France
| | - David Ternant
- Université de Tours, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours, France
| |
Collapse
|
19
|
Leven C, Padelli M, Carré JL, Bellissant E, Misery L. Immune Checkpoint Inhibitors in Melanoma: A Review of Pharmacokinetics and Exposure-Response Relationships. Clin Pharmacokinet 2020; 58:1393-1405. [PMID: 31183812 DOI: 10.1007/s40262-019-00789-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Immune checkpoint inhibitors are a new class of monoclonal antibodies that amplify T-cell-mediated immune responses against cancer cells. The introduction of these new drugs, first anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA4) and then anti-programmed death-1 (anti-PD1), was a major improvement in the treatment of advanced or metastatic melanoma, a highly immunogenic tumour. The development strategy for immune checkpoint immunotherapies differed from that traditionally used for cytotoxic therapies in oncology. The choices of doses at which to conduct clinical trials, and subsequently the choice of doses at which to use these new therapies, were not based on the identification of a maximum tolerated dose from dose-escalation studies; thus, pharmacokinetic and pharmacokinetic-pharmacodynamic modelling was essential. The studies conducted have shown that the pharmacokinetics of ipilimumab were linear and not time-dependent. In addition, there was a correlation between the trough concentrations of ipilimumab and its therapeutic efficacy. On the contrary, the anti-PD1 immunotherapies nivolumab and pembrolizumab had time-dependent pharmacokinetics. Their therapeutic efficacy was not related to their trough concentration, but there was a correlation between the clearance of anti-PD1 and the survival of melanoma patients. This review highlights the complexity of interpreting the exposure-response relationships of these agents. Further studies are needed to assess the value of therapeutic drug monitoring of immune checkpoint inhibitors in the treatment of melanoma.
Collapse
Affiliation(s)
- Cyril Leven
- Department of Biochemistry and Pharmaco-Toxicology, Brest University Hospital, Brest, France. .,University of Brest, LIEN, Brest, France.
| | - Maël Padelli
- Department of Biochemistry and Pharmaco-Toxicology, Brest University Hospital, Brest, France.,University of Brest, LIEN, Brest, France
| | - Jean-Luc Carré
- Department of Biochemistry and Pharmaco-Toxicology, Brest University Hospital, Brest, France.,University of Brest, LIEN, Brest, France
| | - Eric Bellissant
- Department of Clinical and Biological Pharmacology and Pharmacovigilance, Pharmacoepidemiology and Drug Information Centre, Rennes University Hospital, Rennes, France.,Laboratory of Experimental and Clinical Pharmacology, Faculty of Medicine, Rennes 1 University, Rennes, France.,Clinical Investigation Centre, CIC Inserm 1414, Rennes, France
| | - Laurent Misery
- University of Brest, LIEN, Brest, France.,Department of Dermatology, Brest University Hospital, Brest, France
| |
Collapse
|
20
|
Bensalem A, Ternant D. Pharmacokinetic Variability of Therapeutic Antibodies in Humans: A Comprehensive Review of Population Pharmacokinetic Modeling Publications. Clin Pharmacokinet 2020; 59:857-874. [DOI: 10.1007/s40262-020-00874-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Atiqi S, Hooijberg F, Loeff FC, Rispens T, Wolbink GJ. Immunogenicity of TNF-Inhibitors. Front Immunol 2020; 11:312. [PMID: 32174918 PMCID: PMC7055461 DOI: 10.3389/fimmu.2020.00312] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/07/2020] [Indexed: 01/19/2023] Open
Abstract
Tumor necrosis factor inhibitors (TNFi) have significantly improved treatment outcome of rheumatic diseases since their incorporation into treatment protocols two decades ago. Nevertheless, a substantial fraction of patients experiences either primary or secondary failure to TNFi due to ineffectiveness of the drug or adverse reactions. Secondary failure and adverse events can be related to the development of anti-drug antibodies (ADA). The earliest studies that reported ADA toward TNFi mainly used drug-sensitive assays. Retrospectively, we recognize this has led to an underestimation of the amount of ADA produced due to drug interference. Drug-tolerant ADA assays also detect ADA in the presence of drug, which has contributed to the currently reported higher incidence of ADA development. Comprehension and awareness of the assay format used for ADA detection is thus essential to interpret ADA measurements correctly. In addition, a concurrent drug level measurement is informative as it may provide insight in the extent of underestimation of ADA levels and improves understanding the clinical consequences of ADA formation. The clinical effects are dependent on the ratio between the amount of drug that is neutralized by ADA and the amount of unbound drug. Pharmacokinetic modeling might be useful in this context. The ADA response generally gives rise to high affinity IgG antibodies, but this response will differ between patients. Some patients will not reach the phase of affinity maturation while others generate an enduring high titer high affinity IgG response. This response can be transient in some patients, indicating a mechanism of tolerance induction or B-cell anergy. In this review several different aspects of the ADA response toward TNFi will be discussed. It will highlight the ADA assays, characteristics and regulation of the ADA response, impact of immunogenicity on the pharmacokinetics of TNFi, clinical implications of ADA formation, and possible mitigation strategies.
Collapse
Affiliation(s)
- Sadaf Atiqi
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, Netherlands
| | - Femke Hooijberg
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, Netherlands
| | - Floris C Loeff
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Amsterdam, Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Amsterdam, Netherlands
| | - Gerrit J Wolbink
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, Netherlands.,Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Amsterdam, Netherlands
| |
Collapse
|
22
|
Jiang S, Madrasi K, Samant T, Lagishetty C, Vozmediano V, Chiew A, Abdel-Rahman SM, James LP, Schmidt S. Population Pharmacokinetic Modeling of Acetaminophen Protein Adducts in Adults and Children. J Clin Pharmacol 2019; 60:595-604. [PMID: 31802503 DOI: 10.1002/jcph.1555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/24/2019] [Indexed: 11/07/2022]
Abstract
Acetaminophen protein adducts (adducts) are a well-established biomarker to diagnose acetaminophen toxicity. To date, the quantitative relationship between acetaminophen exposure, which drives adduct formation, and adduct exposure remains to be established. Our study characterized the adduct formation and disposition in adults using the approach of population parent-metabolite modeling. It demonstrated formation-limited pharmacokinetics (PK) for adducts in healthy subjects. This finding expands the existing knowledge on adduct PK that showed an apparent long elimination half-life. We then allometrically scaled the adduct PK model to children, simulated the adduct profiles, and compared these simulated profiles with those observed in an independent cohort of children. The scaled model significantly overpredicted the adduct concentrations in children early on in treatment and underpredicted concentrations following repeated acetaminophen doses. These results suggest that children demonstrate different adduct PK behavior from that of adults, most likely because of increased reactive metabolite detoxification in children. In summary, we described the first PK model linking acetaminophen and acetaminophen protein adduct concentrations, which provides a semimechanistic understanding of varying profiles of adduct exposure in adults and children.
Collapse
Affiliation(s)
- Sibo Jiang
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, Florida, USA
| | - Kumpal Madrasi
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, Florida, USA
| | - Tanay Samant
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, Florida, USA
| | - Chakradhar Lagishetty
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, Florida, USA
| | - Valvanera Vozmediano
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, Florida, USA
| | - Angela Chiew
- Department of Clinical Toxicology Prince of Wales Hospital, Randwick, NSW, Australia.,NSW Poisons Information Centre, Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Susan M Abdel-Rahman
- Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospitals and Clinics, Kansas City, Missouri, USA
| | - Laura P James
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, USA
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, Florida, USA
| |
Collapse
|
23
|
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]
|
24
|
Ternant D, Monjanel H, Venel Y, Prunier-Aesch C, Arbion F, Colombat P, Paintaud G, Gyan E. Nonlinear pharmacokinetics of rituximab in non-Hodgkin lymphomas: A pilot study. Br J Clin Pharmacol 2019; 85:2002-2010. [PMID: 31112622 DOI: 10.1111/bcp.13991] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/10/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022] Open
Abstract
AIMS Rituximab is an anti-CD20 monoclonal antibody approved in non-Hodgkin lymphoma (NHL). This study aimed to assess the relationship between antigen mass and nonlinear pharmacokinetics of rituximab in NHL patients. METHODS In a retrospective cohort of 25 NHL patients treated with rituximab, antigen mass was assessed at baseline by measuring metabolic tumour volume (MTV) by positron emission tomography. Rituximab pharmacokinetics was described using a semimechanistic 2-compartment model including a latent target antigen. Rituximab target-mediated elimination was described as irreversible binding between rituximab and it target. Histology (follicular or diffuse large B-cell lymphomas), initial MTV and body weight were tested as covariates on pharmacokinetic parameters. RESULTS The model allowed a satisfactory description of rituximab serum concentrations. Target-mediated elimination was maximum at the beginning of treatment and became negligible towards the end of follow-up. The second-order elimination of rituximab due to target binding and complex elimination increased with baseline MTV. Central volume of distribution increased with body weight (P = .022) and baseline MTV (P = .005). CONCLUSIONS This study quantified for the first time the target-mediated elimination of rituximab in NHL patients and confirmed rituximab retention by antigen mass.
Collapse
Affiliation(s)
- David Ternant
- EA 7501 GICC, Université de Tours, Tours, France.,Service de Pharmacologie Médicale, CHRU de Tours, Tours, France
| | - Hélène Monjanel
- Hématologie et Thérapie Cellulaire, CHRU de Tours, Tours, France
| | - Yann Venel
- Médecine Nucléaire, CHRU de Tours, France
| | | | - Flavie Arbion
- Service d'Anatomie et Cytologie Pathologiques, CHRU de Tours, Tours, France
| | - Philippe Colombat
- Hématologie et Thérapie Cellulaire, CHRU de Tours, Tours, France.,ERL CNRS 7001 LNOx, Université de Tours, Tours, France
| | - Gilles Paintaud
- EA 7501 GICC, Université de Tours, Tours, France.,Service de Pharmacologie Médicale, CHRU de Tours, Tours, France
| | - Emmanuel Gyan
- Hématologie et Thérapie Cellulaire, CHRU de Tours, Tours, France.,ERL CNRS 7001 LNOx, Université de Tours, Tours, France
| |
Collapse
|
25
|
Le Louedec F, Alix-Panabières C, Lafont T, Allal BC, Garrel R, Digue L, Guigay J, Cupissol D, Delord JP, Lallemant B, Alfonsi M, Aubry K, Mazel M, Becher F, Perriard F, Chatelut E, Thomas F. Cetuximab pharmacokinetic/pharmacodynamics relationships in advanced head and neck carcinoma patients. Br J Clin Pharmacol 2019; 85:1357-1366. [PMID: 30811063 DOI: 10.1111/bcp.13907] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/01/2019] [Accepted: 02/16/2019] [Indexed: 12/28/2022] Open
Abstract
AIMS Cetuximab associated with cisplatin and 5-fluorouracil is used to treat patients with inoperable or metastatic head and neck squamous cell carcinomas (HNSCC) up until disease progression or unacceptable toxicities. To date, no biomarkers of efficacy are available to select patients who will benefit from treatment. METHODS An ancillary pharmacokinetics (PK) exploration was performed in the context of a prospective study investigating circulating-tumour cells vs progression-free survival (PFS). Cetuximab plasma concentrations were analysed according to a population PK model. Individual exposure parameters were confronted with soluble epidermal growth factor receptor (sEGFR) concentrations, tumour response and PFS. RESULTS PK data (28 patients, 203 observations) were best described by a two-compartment model with linear elimination. Performance status (PS) significantly correlated to both cetuximab clearance and central volume of distribution with both parameters increasing by 33.3% (95% CI 1-65.6) for each 1-point increase of PS compared to PS = 0. Univariate analysis showed that patients with higher trough cetuximab concentrations at Day 7 (Cmin,D7 ) had better tumour response (P = 0.03) and longer PFS (P = 0.035). However, multivariate analysis revealed that only PS and tumour size at baseline remained significantly associated with PFS. Levels of sEGFR increased during cetuximab treatment but were not associated with PFS in the multivariate analysis. CONCLUSIONS Our study prospectively indicates that PS is likely a confounding factor in the relationship between cetuximab PK and PFS, patients with a poor PS having lower cetuximab plasma exposure and lower PFS.
Collapse
Affiliation(s)
- Félicien Le Louedec
- Laboratory of Pharmacology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France.,CRCT, University of Toulouse, Inserm, Toulouse, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, Montpellier, France
| | - Thierry Lafont
- Laboratory of Pharmacology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France.,CRCT, University of Toulouse, Inserm, Toulouse, France
| | - Ben C Allal
- Laboratory of Pharmacology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France.,CRCT, University of Toulouse, Inserm, Toulouse, France
| | - Renaud Garrel
- Department of Head Neck Cancer and Laryngology, University Medical Centre of Montpellier, France
| | - Laurence Digue
- Medical Oncology Department, CHU Saint André, Bordeaux, France
| | - Joël Guigay
- Department of Head and Neck Oncology, Gustave Roussy, Villejuif, France.,Department of Medical Oncology, Nice, France
| | - Didier Cupissol
- Medical Oncology Department, Institute of Cancer of Montpellier, France
| | - Jean-Pierre Delord
- CRCT, University of Toulouse, Inserm, Toulouse, France.,Medical Oncology Department, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
| | - Benjamin Lallemant
- Department of Head and Neck Surgery, University Hospital Center of Nîmes, France
| | - Marc Alfonsi
- Department of Radiation Oncology, Clinique Sainte Catherine, Avignon, France
| | - Karine Aubry
- Department of Head and Neck Oncology and Surgery, Hôpital Dupuytren CHU, Limoges, France
| | - Martine Mazel
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, Montpellier, France
| | - François Becher
- Service de Pharmacologie et Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Institut National de la Recherche Agronomique, Université Paris Saclay, Gif-sur-Yvette, France
| | - Françoise Perriard
- Department of Biostatistics, UPRES EA2415, Clinical Research University Institute, Montpellier, France
| | - Etienne Chatelut
- Laboratory of Pharmacology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France.,CRCT, University of Toulouse, Inserm, Toulouse, France
| | - Fabienne Thomas
- Laboratory of Pharmacology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France.,CRCT, University of Toulouse, Inserm, Toulouse, France
| |
Collapse
|
26
|
Efficacy of immunotherapy, gut microbiota and impact of antibiotic use: are there confounding factors? Cancer Chemother Pharmacol 2019; 84:223-224. [PMID: 30980131 DOI: 10.1007/s00280-019-03833-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/03/2019] [Indexed: 01/20/2023]
|
27
|
Quartino AL, Li H, Kirschbrown WP, Mangat R, Wada DR, Garg A, Jin JY, Lum B. Population pharmacokinetic and covariate analyses of intravenous trastuzumab (Herceptin ®), a HER2-targeted monoclonal antibody, in patients with a variety of solid tumors. Cancer Chemother Pharmacol 2018; 83:329-340. [PMID: 30467591 PMCID: PMC6394489 DOI: 10.1007/s00280-018-3728-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE The aim of the study was to characterize the population pharmacokinetics (PK) of the intravenous formulation of trastuzumab, assess the impact of patient and pathological covariates on trastuzumab PK, and perform simulations to support dosing recommendations in special situations. METHODS Serum trastuzumab concentrations were obtained from 1582 patients with metastatic breast cancer (MBC), early breast cancer (EBC), advanced gastric cancer (AGC), or other tumor types/healthy volunteers in 18 phase I, II, and III trials and analyzed by nonlinear mixed-effects modeling. RESULTS A two-compartment model with parallel linear and nonlinear elimination best described the data. During treatment, linear clearance (CL) dominated, resulting in a total CL of 0.173-0.337 L/day, which is similar to other IgG1 monoclonal antibodies. Covariates influencing CL were baseline body weight, aspartate aminotransferase, albumin, gastric cancer, and the presence of liver metastases. MBC and EBC had similar PK parameters, while CL was higher in AGC. Simulations indicated that at least 95% of patients with BC reach concentrations < 1 µg/mL (~ 97% washout) by 7 months. A dose delay in BC or AGC patients of > 1 week would take approximately 6 weeks to get back within steady-state exposure range. CONCLUSIONS Trastuzumab PK for the intravenous formulation was well-described across cancer types, disease status, and regimens. No dose adjustment is required for any of the identified patient covariates. A 7-month serum washout period for trastuzumab is recommended. A reloading dose is required if a maintenance dose is missed by > 1 week.
Collapse
Affiliation(s)
- Angelica L Quartino
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA.
| | - Hanbin Li
- Certara, L.P., 845 Oak Grove Ave, Menlo Park, CA, 94025, USA
| | | | - Ranvir Mangat
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA.,Insight Rx, 233 Stanyan Street, San Francisco, CA, 94118, USA
| | - D Russell Wada
- Certara, L.P., 845 Oak Grove Ave, Menlo Park, CA, 94025, USA
| | - Amit Garg
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA
| | - Jin Y Jin
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA
| | - Bert Lum
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA
| |
Collapse
|