1
|
Guo Y, Remaily BC, Thomas J, Kim K, Kulp SK, Mace TA, Ganesan LP, Owen DH, Coss CC, Phelps MA. Antibody Drug Clearance: An Underexplored Marker of Outcomes with Checkpoint Inhibitors. Clin Cancer Res 2024; 30:942-958. [PMID: 37921739 PMCID: PMC10922515 DOI: 10.1158/1078-0432.ccr-23-1683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/23/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Immune-checkpoint inhibitor (ICI) therapy has dramatically changed the clinical landscape for several cancers, and ICI use continues to expand across many cancer types. Low baseline clearance (CL) and/or a large reduction of CL during treatment correlates with better clinical response and longer survival. Similar phenomena have also been reported with other monoclonal antibodies (mAb) in cancer and other diseases, highlighting a characteristic of mAb clinical pharmacology that is potentially shared among various mAbs and diseases. Though tempting to attribute poor outcomes to low drug exposure and arguably low target engagement due to high CL, such speculation is not supported by the relatively flat exposure-response relationship of most ICIs, where a higher dose or exposure is not likely to provide additional benefit. Instead, an elevated and/or increasing CL could be a surrogate marker of the inherent resistant phenotype that cannot be reversed by maximizing drug exposure. The mechanisms connecting ICI clearance, therapeutic efficacy, and resistance are unclear and likely to be multifactorial. Therefore, to explore the potential of ICI CL as an early marker for efficacy, this review highlights the similarities and differences of CL characteristics and CL-response relationships for all FDA-approved ICIs, and we compare and contrast these to selected non-ICI mAbs. We also discuss underlying mechanisms that potentially link mAb CL with efficacy and highlight existing knowledge gaps and future directions where more clinical and preclinical investigations are warranted to clearly understand the value of baseline and/or time-varying CL in predicting response to ICI-based therapeutics.
Collapse
Affiliation(s)
- Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Bryan C. Remaily
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Justin Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Kyeongmin Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Samuel K. Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Latha P. Ganesan
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Dwight H. Owen
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Mitch A. Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| |
Collapse
|
2
|
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
|
3
|
Reig-Lopez J, Tang W, Fernandez-Teruel C, Merino-Sanjuan M, Mangas-Sanjuan V, Boulton DW, Sharma P. Application of population physiologically based pharmacokinetic modelling to optimize target expression and clearance mechanisms of therapeutic monoclonal antibodies. Br J Clin Pharmacol 2023; 89:2691-2702. [PMID: 37055941 DOI: 10.1111/bcp.15745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/12/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023] Open
Abstract
AIMS To use population physiologically based pharmacokinetic (PopPBPK) modelling to optimize target expression, kinetics and clearance of HER1/2 directed therapeutic monoclonal antibodies (mAbs). Thus, to propose a general workflow of PopPBPK modelling and its application in clinical pharmacology. METHODS Full PBPK model of pertuzumab (PTZ) was developed in patient population using Simcyp V21R1 incorporating mechanistic targeted-mediated drug disposition process by fitting known clinical PK and sparse receptor proteomics data to optimize target expression and kinetics of HER2 receptor. Trastuzumab (TTZ) PBPK modelling was used to validate the optimized HER2 target. Additionally, the simulator was also used to develop a full PBPK model for the HER1-directed mAb cetuximab (CTX) to assess the underlying targeted-mediated drug disposition-independent elimination mechanisms. RESULTS HER2 final parameterisation coming from the PBPK modelling of PTZ was successfully cross validated through PBPK modelling of TTZ with average fold error (AFE), absolute AFE and percent prediction error values for area under the concentration-time curve (AUC) and maximum plasma concentration (Cmax ) of 1.13, 1.16 and 16, and 1.01, 1.07 and 7, respectively. CTX PBPK model performance was validated after the incorporation of an additional systemic clearance of 0.033 L/h as AFE and absolute AFE showed an acceptable predictive power of AUC and Cmax with percent prediction error of 13% for AUC and 10% for Cmax . CONCLUSIONS Optimisation of both system and drug related parameters were performed through PBPK modelling to improve model performance of therapeutic mAbs (PTZ, TTZ and CTX). General workflow was proposed to develop and apply PopPBPK to support clinical development of mAbs targeting same receptor.
Collapse
Affiliation(s)
- Javier Reig-Lopez
- Pharmacy and Pharmaceutical Technology and Parasitology Department, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Weifeng Tang
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Carlos Fernandez-Teruel
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Matilde Merino-Sanjuan
- Pharmacy and Pharmaceutical Technology and Parasitology Department, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, Valencia, Spain
| | - Victor Mangas-Sanjuan
- Pharmacy and Pharmaceutical Technology and Parasitology Department, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, Valencia, Spain
| | - David W Boulton
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Pradeep Sharma
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
4
|
Imai C, Saeki H, Yamamoto K, Ichikawa A, Arai M, Tawada A, Suzuki T, Takiguchi Y, Hanazawa T, Ishii I. Radiotherapy plus cetuximab for locally advanced squamous cell head and neck cancer in patients with cisplatin‑ineligible renal dysfunction: A retrospective study. Oncol Lett 2022; 23:152. [PMID: 35836484 PMCID: PMC9258593 DOI: 10.3892/ol.2022.13271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 11/05/2022] Open
Abstract
Clinical trials have not fully demonstrated the efficacy and safety of radiotherapy plus cetuximab for locally advanced squamous cell head and neck cancer (LA-SCCHN) in patients with cisplatin-ineligible renal dysfunction. Patients who received radiotherapy plus cetuximab for LA-SCCHN at Chiba University Hospital (Chiba, Japan) between July 2013 and October 2018 were retrospectively reviewed. Background characteristics and locoregional control and overall survival rates were compared between patients with and without renal dysfunction. Survival was examined using Kaplan-Meier analysis and an adjusted Cox proportional hazards model. Kaplan-Meier analysis demonstrated that overall survival was shorter in patients with creatinine clearance of <45 ml/min (P=0.041; log-rank test). However, there was no difference in the locoregional control rate (P=0.477; log-rank test). Adjusted Cox analysis revealed that the risk of death was increased by 2.52-fold (hazard ratio, 2.52; 95% confidence interval, 1.01-6.30; P=0.048) if creatinine clearance was <45 ml/min. Moderate to severe renal dysfunction did not affect the locoregional control rate in patients with LA-SCCHN treated with radiotherapy plus cetuximab but was an adverse prognostic factor.
Collapse
Affiliation(s)
- Chiaki Imai
- Division of Pharmacy, Chiba University Hospital, Chiba, Chiba 260‑8677, Japan
| | - Hiromi Saeki
- Division of Pharmacy, Chiba University Hospital, Chiba, Chiba 260‑8677, Japan
| | - Kohei Yamamoto
- Division of Pharmacy, Chiba University Hospital, Chiba, Chiba 260‑8677, Japan
| | - Ayano Ichikawa
- Division of Pharmacy, Chiba University Hospital, Chiba, Chiba 260‑8677, Japan
| | - Makoto Arai
- Department of Gastroenterology, Tokyo Women's Medical University, Yachiyo Medical Center, Yachiyo, Chiba 276‑8524, Japan
| | - Akinobu Tawada
- Department of Nursing, Faculty of Health Care Sciences, Chiba Prefectural University of Health Sciences, Chiba, Chiba 261‑0014, Japan
| | - Takaaki Suzuki
- Division of Pharmacy, Chiba University Hospital, Chiba, Chiba 260‑8677, Japan
| | - Yuichi Takiguchi
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chiba, Chiba 260‑8670, Japan
| | - Toyoyuki Hanazawa
- Department of Otorhinolaryngology/Head and Neck Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba 260‑8670, Japan
| | - Itsuko Ishii
- Division of Pharmacy, Chiba University Hospital, Chiba, Chiba 260‑8677, Japan
| |
Collapse
|
5
|
Population pharmacokinetics and exposure-response relationship of trastuzumab and bevacizumab in early-stage breast cancer. Eur J Clin Pharmacol 2021; 77:1861-1873. [PMID: 34245336 DOI: 10.1007/s00228-021-03179-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 06/21/2021] [Indexed: 10/20/2022]
Abstract
AIMS To describe the sources of interindividual variability of bevacizumab and trastuzumab pharmacokinetics in early-stage breast cancer, and to study the relationship between exposure and both early clinical response and specific adverse events. PATIENTS AND METHODS Patients (n = 86) received 6 cycles of docetaxel + trastuzumab. Early tumour response was assessed by determination of the maximum standard uptake value (SUVmax) variation (ΔSUVmax) after 1 cycle using [18F]-fluorodeoxyglucose (FDG) PET. Early poor responders (ΔSUVmax < 70%) also received bevacizumab from cycle 3 to cycle 6. Sources of interindividual variability in pharmacokinetics of both antibodies were studied by population compartment modelling. Exposure as assessed by area under the concentration-versus-time curve (AUC) was compared between responders and non-responders and between patients experiencing specific adverse events or not. RESULTS A two-compartment model described the pharmacokinetics of both antibodies satisfactorily. Their central volume of distributions (Vc) increased with body surface area and their elimination half-lives were shorter (~14 days) than previously reported (~26-28 days). There was a time-dependent increase in trastuzumab Vc, positively correlated to baseline SUVmax. Bevacizumab elimination rate (k10) was positively correlated with ΔSUVmax measured at the end of the first cycle. Bevacizumab had no significantly influence on trastuzumab pharmacokinetics. No relationship between exposure and clinical response or occurrence of adverse events was found. CONCLUSION Tumour uptake as assessed by SUVmax influences the pharmacokinetics of bevacizumab and trastuzumab. In early-stage breast cancer, elimination half-lives of these therapeutic monoclonal antibodies may be shorter than those previously reported in more advanced disease.
Collapse
|
6
|
Kamashev D, Sorokin M, Kochergina I, Drobyshev A, Vladimirova U, Zolotovskaia M, Vorotnikov I, Shaban N, Raevskiy M, Kuzmin D, Buzdin A. Human blood serum can donor-specifically antagonize effects of EGFR-targeted drugs on squamous carcinoma cell growth. Heliyon 2021; 7:e06394. [PMID: 33748471 PMCID: PMC7966997 DOI: 10.1016/j.heliyon.2021.e06394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/29/2020] [Accepted: 02/25/2021] [Indexed: 02/09/2023] Open
Abstract
Many patients fail to respond to EGFR-targeted therapeutics, and personalized diagnostics is needed to identify putative responders. We investigated 1630 colorectal and lung squamous carcinomas and 1357 normal lung and colon samples and observed huge variation in EGFR pathway activation in both cancerous and healthy tissues, irrespectively on EGFR gene mutation status. We investigated whether human blood serum can affect squamous carcinoma cell growth and EGFR drug response. We demonstrate that human serum antagonizes the effects of EGFR-targeted drugs erlotinib and cetuximab on A431 squamous carcinoma cells by increasing IC50 by about 2- and 20-fold, respectively. The effects on clonogenicity varied significantly across the individual serum samples in every experiment, with up to 100% differences. EGF concentration could explain many effects of blood serum samples, and EGFR ligands-depleted serum showed lesser effect on drug sensitivity.
Collapse
Affiliation(s)
- Dmitry Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
| | - Maksim Sorokin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Irina Kochergina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
| | - Aleksey Drobyshev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Uliana Vladimirova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
| | - Marianna Zolotovskaia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Igor Vorotnikov
- Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| | - Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Mikhail Raevskiy
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
- OmicsWay Corp., Walnut, CA, USA
| | - Denis Kuzmin
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| |
Collapse
|
7
|
Correlations between serum cetuximab and EGFR-related markers, and skin disorders in head and neck cancer patients. Cancer Chemother Pharmacol 2021; 87:555-565. [PMID: 33462734 DOI: 10.1007/s00280-020-04228-4] [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: 09/02/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Cetuximab inhibits epidermal growth factor receptor (EGFR) signaling in cancer and skin cells, thereby inducing anti-cancer effects and skin disorders. The present study aimed to evaluate the relationships between serum cetuximab and EGFR-related markers, and adverse effects in head and neck cancer patients. METHODS Thirty-four head and neck cancer patients receiving weekly intravenous cetuximab were enrolled. Serum cetuximab levels were determined just before dosing. Blood samples for determination of serum EGFR-related markers including soluble epidermal growth factor receptor (sEGFR) and interleukin-6 (IL-6) were obtained. The severities of skin disorders, their medications, and hypomagnesemia treatment were also assessed. RESULTS Serum levels of cetuximab and sEGFR were negatively and positively correlated with that of IL-6, respectively. The serum cetuximab level was twofold higher in the patients with a grade 2-3 skin rash than with a grade 0-1 rash. The serum cetuximab cutoff value related to severe skin rash was 71 μg/mL (sensitivity, 59%; and specificity, 94%). The use of a strong topical corticosteroid for skin rash was also associated with a higher serum cetuximab level. Serum levels of sEGFR and IL-6 had no correlations with the skin disorder severities or their medications. Hypomagnesemia treatment using intravenous magnesium sulfate was not related to serum cetuximab and EGFR-related markers. CONCLUSIONS Head and neck cancer patients with a higher serum IL-6 level tended to have a lower serum cetuximab level. Serum cetuximab had positive correlations to skin rash severity and its medication in the study population.
Collapse
|
8
|
Kuilamu E, Subasic C, Cowin GJ, Simpson F, Minchin RF, Kaminskas LM. Cetuximab Exhibits Sex Differences in Lymphatic Exposure after Intravenous Administration in Rats in the Absence of Differences in Plasma Exposure. Pharm Res 2020; 37:224. [PMID: 33078255 DOI: 10.1007/s11095-020-02945-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/05/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of this work was to identify whether biochemical and physiological sources of mAb pharmacokinetic sex-effects could be identified in the rat model where target-mediated disposition is avoided. METHODS Plasma and lymphatic pharmacokinetics of the humanised anti-EGFR antibody cetuximab, along with potential physiological and biochemical drivers of pharmacokinetic sex differences, were examined in male and female rats. Cetuximab was used as a model mAb since plasma clearance is slower in female patients. RESULTS When plasma concentrations were normalised to dose, female rats displayed slower plasma clearance than males, but no significant differences were observed in liver and spleen biodistribution. Sex differences in apparent plasma clearance, however, were abolished after normalisation to body weight, surface area or fat-free mass. Significant sex differences were observed in plasma testosterone, endogenous IgG and fat free mass, but did not correlate with apparent clearance. Females did, however, show two-fold higher lymphatic exposure compared to males. CONCLUSIONS These data suggested that mAbs more efficiently access lymph in females, but this does not affect plasma pharmacokinetics or biodistribution. Further, the data suggest that sex differences observed in humans could be a function of antigen density.
Collapse
Affiliation(s)
- Esther Kuilamu
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Christopher Subasic
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Gary J Cowin
- National Imaging Facility, Centre for Advanced Imaging, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Fiona Simpson
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Lisa M Kaminskas
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
9
|
Grisic AM, Khandelwal A, Bertolino M, Huisinga W, Girard P, Kloft C. Semimechanistic Clearance Models of Oncology Biotherapeutics and Impact of Study Design: Cetuximab as a Case Study. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:628-638. [PMID: 33015996 PMCID: PMC7679074 DOI: 10.1002/psp4.12558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022]
Abstract
This study aimed to explore the currently competing and new semimechanistic clearance models for monoclonal antibodies and the impact of clearance model misspecification on exposure metrics under different study designs exemplified for cetuximab. Six clearance models were investigated under four different study designs (sampling density and single/multiple‐dose levels) using a rich data set from two cetuximab clinical trials (226 patients with metastatic colorectal cancer) and using the nonlinear mixed‐effects modeling approach. A two‐compartment model with parallel Michaelis–Menten and time‐decreasing linear clearance adequately described the data, the latter being related to post‐treatment response. With respect to bias in exposure metrics, the simplified time‐varying linear clearance (CL) model was the best alternative. Time‐variance of the linear CL component should be considered for biotherapeutics if response impacts pharmacokinetics. Rich sampling at steady‐state was crucial for unbiased estimation of Michaelis–Menten elimination in case of the reference (parallel Michaelis–Menten and time‐varying linear CL) model.
Collapse
Affiliation(s)
- Ana-Marija Grisic
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany.,Graduate Research Training Program PharMetrX, Berlin, Germany.,Merck KGaA, Darmstadt, Germany
| | | | | | | | - Pascal Girard
- Merck Institute of Pharmacometrics, Merck Serono S.A., Lausanne, Switzerland
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| |
Collapse
|
10
|
Xiang H, Liu L, Gao Y, Ahene A, Macal M, Hsu AW, Dreiling L, Collins H. Population pharmacokinetic analysis of phase 1 bemarituzumab data to support phase 2 gastroesophageal adenocarcinoma FIGHT trial. Cancer Chemother Pharmacol 2020; 86:595-606. [PMID: 32965540 PMCID: PMC7561547 DOI: 10.1007/s00280-020-04139-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/06/2020] [Indexed: 12/11/2022]
Abstract
Purpose To report population pharmacokinetic (PK) analysis of the phase 1 study (FPA144-001, NCT02318329) and to select a clinical dose and schedule that will achieve an empirical target trough concentration (Ctrough) for an anti-fibroblast growth factor receptor 2b antibody, bemarituzumab. Methods Nonlinear mixed-effect modeling was used to analyse PK data. In vitro binding affinity and receptor occupancy of bemarituzumab were determined. Simulation was conducted to estimate dose and schedule to achieve an empirical target Ctrough in a phase 2 trial (FIGHT, NCT03694522) for patients receiving first-line treatment combined with modified 5-fluourouracil, oxaliplatin and leucovorin (mFOLFOX6) for gastric and gastroesophageal junction adenocarcinoma. Results Bemarituzumab PK is best described by a two-compartment model with parallel linear and nonlinear (Michaelis–Menten) elimination from the central compartment. Albumin, gender, and body weight were identified as the covariates on the linear clearance and/or volume of distribution in the central compartment, and no dose adjustment was warranted. An empirical target of bemarituzumab Ctrough of ≥ 60 µg/mL was projected to achieve > 95% receptor occupancy based on in vitro data. Fifteen mg/kg every 2 weeks, with a single dose of 7.5 mg/kg on Cycle 1 Day 8, was projected to achieve the target Ctrough on Day 15 in 98% of patients with 96% maintaining the target at steady state, which was confirmed in the FIGHT trial. Conclusion A projected dose and schedule to achieve the target Ctrough was validated in phase 1 of the FIGHT trial which supported selection of the phase 2 dose and schedule for bemarituzumab. Electronic supplementary material The online version of this article (10.1007/s00280-020-04139-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Xiang
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA.
| | - Lucy Liu
- Shanghai Qiangshi Information Technology Co., Ltd, Shanghai, China
| | - Yuying Gao
- Shanghai Qiangshi Information Technology Co., Ltd, Shanghai, China
| | - Ago Ahene
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Monica Macal
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA.,TRex Bio, Inc., South San Francisco, CA, USA
| | - Amy W Hsu
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA.,Merck and Co., South San Francisco, CA, USA
| | - Lyndah Dreiling
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Helen Collins
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| |
Collapse
|
11
|
Ternant D, Azzopardi N, Raoul W, Bejan-Angoulvant T, Paintaud G. Influence of Antigen Mass on the Pharmacokinetics of Therapeutic Antibodies in Humans. Clin Pharmacokinet 2020; 58:169-187. [PMID: 29802542 DOI: 10.1007/s40262-018-0680-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic antibodies are increasingly used to treat various diseases, including neoplasms and chronic inflammatory diseases. Antibodies exhibit complex pharmacokinetic properties, notably owing to the influence of antigen mass, i.e. the amount of antigenic targets to which the monoclonal antibody binds specifically. This review focuses on the influence of antigen mass on the pharmacokinetics of therapeutic antibodies quantified by pharmacokinetic modelling in humans. Out of 159 pharmacokinetic studies, 85 reported an influence of antigen mass. This influence led to non-linear elimination decay in 50 publications, which was described using target-mediated drug disposition or derived models, as quasi-steady-state, irreversible binding and Michaelis-Menten models. In 35 publications, the pharmacokinetics was apparently linear and the influence of antigen mass was described as a covariate of pharmacokinetic parameters. If some reported covariates, such as the circulating antigen level or tumour size, are likely to be correlated to antigen mass, others, such as disease activity or disease type, may contain little information on the amount of antigenic targets. In some cases, antigen targets exist in different forms, notably in the circulation and expressed at the cell surface. The influence of antigen mass should be soundly described during the early clinical phases of drug development. To maximise therapeutic efficacy, sufficient antibody doses should be administered to ensure the saturation of antigen targets by therapeutic antibodies in all patients. If necessary, antigen mass should be taken into account in routine clinical practice.
Collapse
Affiliation(s)
- David Ternant
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France. .,Department of Medical Pharmacology, CHRU de Tours, Tours University Hospital, 2 boulevard Tonnellé, 37044, Tours Cedex, France.
| | | | - William Raoul
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France
| | - Theodora Bejan-Angoulvant
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours University Hospital, 2 boulevard Tonnellé, 37044, Tours Cedex, France
| | - Gilles Paintaud
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours University Hospital, 2 boulevard Tonnellé, 37044, Tours Cedex, France
| |
Collapse
|
12
|
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: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Abstract
Cancer immunotherapy is based on checkpoint inhibitors (CPIs) that significantly improve the clinical outcome of several malignant diseases. These inhibitors are monoclonal antibodies (mAbs) directed at cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or programmed death-ligand 1 (PD-L1), sharing most of the clinical pharmacokinetic characteristics of mAb targeted therapies, all of which differ from those of cytotoxics and small molecules. Establishing the labeled dose of mAbs, and particularly of the CPIs, represents a true challenge. This review therefore examines the main criteria used for dose selection, along with their limits. The relationships between CPI pharmacokinetic parameters and treatment outcome (efficacy and/or toxicity) differ somewhat among the various drugs, but general features can be identified. Nevertheless, the interpretation of these relationships remains quite controversial. A first interpretation asserts that inter-individual pharmacokinetic variability in clearance has an impact on outcome and should be taken into consideration for dosing individualization. The second considers that higher clearance values observed in some patients result from characteristics associated with poor predictive factors of efficacy. Finally, the schedule, and particularly its frequency of administration, merits rethinking.
Collapse
|
14
|
Pharmacokinetics of Anticancer Drugs Used in Treatment of Older Adults With Colorectal Cancer: A Systematic Review. Ther Drug Monit 2019; 41:553-560. [DOI: 10.1097/ftd.0000000000000635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
15
|
Bajaj G, Suryawanshi S, Roy A, Gupta M. Evaluation of covariate effects on pharmacokinetics of monoclonal antibodies in oncology. Br J Clin Pharmacol 2019; 85:2045-2058. [PMID: 31140642 DOI: 10.1111/bcp.13996] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/25/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
AIMS The development of monoclonal antibodies (mAbs) requires an understanding of the interindividual variability (IIV) in pharmacokinetics (PK) at the population level facilitated by population PK (PopPK) modelling. However, there is no clear rationale for selecting which covariates to screen during PopPK model development. Here, we compare the effect of covariates on PK parameters for mAbs in oncology and identify the most commonly used covariates affecting PK parameters. METHODS All 25 mAbs approved for therapeutic use in oncology until December 2017 by the Food and Drug Administration and the European Medicines Agency were selected for study. Literature searches revealed 23 available PopPK models for these mAbs. To understand the magnitude and types of covariate effect on PK parameters, all covariates included in the final PopPK model for each mAb were summarized. RESULTS The most commonly identified covariates were baseline body weight (BW; 17 mAbs), baseline serum albumin (8 mAbs), and sex (7 mAbs) on clearance; and BW (16 mAbs) and sex (12 mAbs) on central volume of distribution. A reduced PopPK model was developed for nivolumab and ipilimumab using these covariates, and the percentage of explained IIV from the reduced model (20.3% and 16.8%, respectively) was compared with that from the full model (24.5% and 27.9%, respectively). CONCLUSIONS This analysis provides a uniform platform for selecting covariates and suggests that the effect of BW, albumin and sex should be included during the development of PopPK models for mAbs in oncology. The reduced model was able to explain IIV to a similar extent as the full model.
Collapse
Affiliation(s)
| | | | - Amit Roy
- Bristol-Myers Squibb, Princeton, NJ, USA
| | | |
Collapse
|
16
|
Clinical Pharmacokinetics and Pharmacodynamics of the Epidermal Growth Factor Receptor Inhibitor Panitumumab in the Treatment of Colorectal Cancer. Clin Pharmacokinet 2019; 57:455-473. [PMID: 28853050 PMCID: PMC5856878 DOI: 10.1007/s40262-017-0590-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite progress in the treatment of metastatic colorectal cancer (mCRC) in the last 15 years, it is still a condition with a relatively low 5-year survival rate. Panitumumab, a fully human monoclonal antibody directed against the epidermal growth factor receptor (EGFR), is able to prolong survival in patients with mCRC. Panitumumab is used in different lines of therapy in combination with chemotherapy, and as monotherapy for the treatment of wild-type (WT) RAS mCRC. It is administered as an intravenous infusion of 6 mg/kg every 2 weeks and has a t½ of approximately 7.5 days. Elimination takes place via two different mechanisms, and immunogenicity rates are low. Only RAS mutations have been confirmed as a negative predictor of efficacy with anti-EGFR antibodies. Panitumumab is generally well tolerated and has a manageable toxicity profile, despite a very high prevalence of dermatologic side effects. This article presents an overview of the clinical pharmacokinetics and pharmacodynamics of panitumumab, including a description of the studies that led to its approval in the different lines of therapy of mCRC.
Collapse
|
17
|
Ogungbenro K, Patel A, Saunders M, Clark J, Duncombe R. An evaluation of cetuximab dosing strategies using pharmacokinetics and cost analysis. ACTA ACUST UNITED AC 2019; 71:1222-1230. [PMID: 31124587 DOI: 10.1111/jphp.13108] [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: 09/27/2018] [Accepted: 05/05/2019] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Cetuximab dosing is based on body surface area (BSA), an approach that is associated with significant wastage due to available vial sizes. NHS England recently introduced an alternative strategy for cetuximab dosing based on dose banding. The aim of this work was to investigate approaches to cetuximab dosing to improve its cost-effectiveness. METHODS A simulation study using a population pharmacokinetic model was used to assess the performance of dosing strategies using exposure, probability of target attainment and cost. Two dosage regimens (500 and 400/250 mg/m2 ) were investigated; 5% and 10% dose banding, fixed and optimised dosing strategies were evaluated and compared to BSA strategy. KEY FINDINGS The percentage of the total cost associated with wastage for the 400/250 mg/m2 regimen were 8.75%, 5.13%, 3.61%, 9.2% and 0% for BSA; 5 and 10% bands; fixed and optimal strategies, respectively. Similar results were obtained for 500 mg/m2 regimen. In comparison with BSA strategy, other strategies have comparable or improved performance. Optimised strategy showed consistent performance and ensures equal exposure and probability of target attainment. CONCLUSIONS Cost-effectiveness of cetuximab treatment can be improved with alternative strategies by reducing wastage without compromising exposure.
Collapse
Affiliation(s)
- Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,The Christie NHS Foundation Trust, Manchester, UK
| | - Alkesh Patel
- The Christie NHS Foundation Trust, Manchester, UK
| | | | - James Clark
- The Christie NHS Foundation Trust, Manchester, UK
| | | |
Collapse
|
18
|
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: 3.6] [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
|
19
|
de Castro-Suárez N, Rodríguez-Vera L, Villegas C, Dávalos-Iglesias JM, Bacallao-Mendez R, Llerena-Ferrer B, Leyva-de la Torre C, Lorenzo-Luaces P, Troche-Concepción M, Ramos-Suzarte M. Pharmacokinetic Evaluation of Nimotuzumab in Patients With Autosomal Dominant Polycystic Kidney Disease. J Clin Pharmacol 2019; 59:863-871. [PMID: 30633365 DOI: 10.1002/jcph.1376] [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: 06/13/2018] [Accepted: 12/19/2018] [Indexed: 01/05/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disease characterized by an overexpression and mislocalization of epidermal growth factor receptor (EGFR) to the apical membranes of cystic epithelial cells. Nimotuzumab is a humanized antibody that recognizes an extracellular domain III of human EGFR. The aim of this study was to assess the pharmacokinetic behavior of nimotuzumab in patients with ADPKD given as a single dose. A phase I, single-center, and noncontrolled open clinical study was conducted. Five patients were enrolled at each of the following fixed-dose levels: 50, 100, 200, and 400 mg. Intravenous continuous infusions of nimotuzumab were administered every 14 days during a year, except the first administration, when blood samples were drawn during 28 days for pharmacokinetic assessments. Subjects were closely monitored during the trial and at completion of the administration of nimotuzumab, including the anti-idiotypic response. For the first time, nimotuzumab was used for treating a nononcological disease. The administration of nimotuzumab showed dose-dependent kinetics. Nimotuzumab does not develop anti-idiotypic response against the murine portion present in the hypervariable region of the antibody present in the serum of the patients treated. No significant differences were found in the systemic clearance between the 100- and 400-mg dose, which indicates that the optimal biological dose is in this range of dose.
Collapse
Affiliation(s)
- Niurys de Castro-Suárez
- Laboratory of Biopharmaceutics, Department of Pharmacology & Toxicology, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | - Leyanis Rodríguez-Vera
- Laboratory of Biopharmaceutics, Department of Pharmacology & Toxicology, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | - Carlos Villegas
- National Institute of Oncology and Radiobiology, Havana, Cuba
| | | | | | | | | | | | | | | |
Collapse
|
20
|
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: 20] [Impact Index Per Article: 3.3] [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
|
21
|
Abstract
Necitumumab is a second-generation, recombinant, human immunoglobulin G1, epidermal growth factor (EGFR) receptor antibody that specifically blocks the ligand binding site of EGFR. Necitumumab potentially acts by blocking ligand epidermal growth factor (EGF) binding-mediated activation of the EGFR signaling pathway, inhibiting tumor growth, angiogenesis, and anti-apoptotic mechanisms. Necitumumab inhibited the interaction of EGF and EGFR with a concentration that inhibits binding by 50 % of approximately 0.9 nM (0.13 mg/L) and demonstrated antitumor activity during in vivo experiments associated with trough plasma concentrations of approximately 40 mg/L. This work describes the population pharmacokinetics of necitumumab in cancer patients when administered with or without concomitant chemotherapy and evaluates patient characteristics that may guide dosing. Nonlinear mixed-effects modeling of serum concentration data across five clinical studies (phases I–III) indicated that necitumumab exhibited target-mediated drug disposition, commonly observed with monoclonal antibodies, and that pharmacokinetics were expected to be linear in the studied dose ranges when administered as repeated infusions. No age, sex, race, or concomitant medication factors were found influential, while weight was a statistically significant factor for both distribution and elimination. Simulations from the final model indicated that only a limited reduction in patient drug exposure variability would be achieved by weight- or body surface area-based dosing. Necitumumab effective half-life was estimated to approximately 2 weeks, and steady state was achieved within three to four cycles of treatment. The phase III dosing schedule of 800 mg dosed on days 1 and 8 of a 21-day schedule resulted in serum concentrations that exceeded the 40-mg/L threshold indicated by preclinical experiments.
Collapse
Affiliation(s)
- Amanda Long
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Emmanuel Chigutsa
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Johan Wallin
- Lilly Sweden, Gustav III Boulevard 42, P.O box 721, SE - 169 27, Solna, Sweden.
| |
Collapse
|
22
|
Cetuximab Pharmacokinetics Influences Overall Survival in Patients With Head and Neck Cancer. Ther Drug Monit 2017; 38:567-72. [PMID: 27631463 DOI: 10.1097/ftd.0000000000000321] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND A retrospective study was conducted to analyze interindividual variability of cetuximab pharmacokinetics and its influence on survival (progression-free survival and overall survival [OS]) in a cohort of head and neck squamous cell carcinoma (HNSCC). METHODS Thirty-four patients received cetuximab as an infusion loading dose of 400 mg/m followed by weekly infusions of 250 mg/m. Twenty-one patients had locally advanced HNSCC, and 13 had metastatic/recurrent HNSCC. Cetuximab concentrations were measured by the enzyme-linked immunosorbent assay, and its pharmacokinetics was analyzed by a population approach. Survivals were analyzed with the log-rank test. RESULTS Cetuximab pharmacokinetics was best described using a 2-compartment model with both first-order and saturable (zero-order) eliminations. Estimated pharmacokinetic parameters (%CV) were central volume of distribution V1 = 3.18 L (6%), peripheral volume of distribution V2 = 5.4 L (42%), elimination clearance CL = 0.57 L/d (31%), distribution clearance Q = 0.64 L/d, and zero-order elimination rate k0 = 6.72 mg/d (29%). Both V1 and V2 increased with the body surface area. Adjunction of chemotherapy reduced CL and increased k0. OS was inversely related with cetuximab global clearance (P = 0.007) and was higher in patients with severe radiation dermatitis (P = 0.005). CONCLUSIONS Cetuximab pharmacokinetics in patients with HNSCC can be described using a 2-compartment model combining linear and nonlinear mechanisms of elimination. OS is associated with both cetuximab global clearance and severe radiation dermatitis.
Collapse
|
23
|
Shibata K, Naito T, Okamura J, Hosokawa S, Mineta H, Kawakami J. Simple and rapid LC-MS/MS method for the absolute determination of cetuximab in human serum using an immobilized trypsin. J Pharm Biomed Anal 2017; 146:266-272. [DOI: 10.1016/j.jpba.2017.08.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
|
24
|
Gill KL, Machavaram KK, Rose RH, Chetty M. Potential Sources of Inter-Subject Variability in Monoclonal Antibody Pharmacokinetics. Clin Pharmacokinet 2017; 55:789-805. [PMID: 26818483 DOI: 10.1007/s40262-015-0361-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Understanding inter-subject variability in drug pharmacokinetics and pharmacodynamics is important to ensure that all patients attain suitable drug exposure to achieve efficacy and avoid toxicity. Inter-subject variability in the pharmacokinetics of therapeutic monoclonal antibodies (mAbs) is generally moderate to high; however, the factors responsible for the high inter-subject variability have not been comprehensively reviewed. In this review, the extent of inter-subject variability for mAb pharmacokinetics is presented and potential factors contributing to this variability are explored and summarised. Disease status, age, sex, ethnicity, body size, genetic polymorphisms, concomitant medication, co-morbidities, immune status and multiple other patient-specific details have been considered. The inter-subject variability for mAb pharmacokinetics most likely depends on the complex interplay of multiple factors. However, studies aimed at investigating the reasons for the inter-subject variability are sparse. Population pharmacokinetic models and physiologically based pharmacokinetic models are useful tools to identify important covariates, aiding in the understanding of factors contributing to inter-subject variability. Further understanding of inter-subject variability in pharmacokinetics should aid in development of dosing regimens that are more appropriate.
Collapse
Affiliation(s)
- Katherine L Gill
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Krishna K Machavaram
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Rachel H Rose
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Manoranjenni Chetty
- Simcyp (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK.
| |
Collapse
|
25
|
Abstract
Monoclonal antibodies (MAbs) have become a substantial part of many pharmaceutical company portfolios. However, the development process of MAbs for clinical use is quite different than for small-molecule drugs. MAb development programs require careful interdisciplinary evaluations to ensure the pharmacology of both the MAb and the target antigen are well-understood. Selection of appropriate preclinical species must be carefully considered and the potential development of anti-drug antibodies (ADA) during these early studies can limit the value and complicate the performance and possible duration of preclinical studies. In human studies, many of the typical pharmacology studies such as renal or hepatic impairment evaluations may not be needed but the pharmacokinetics and pharmacodynamics of these agents is complex, often necessitating more comprehensive evaluation of clinical data and more complex bioanalytical assays than might be used for small molecules. This paper outlines concerns and strategies for development of MAbs from the early in vitro assessments needed through preclinical and clinical development. This review focuses on how to develop, submit, and comply with regulatory requirements for MAb therapeutics.
Collapse
|
26
|
Park W, Han S, Lee J, Hong T, Won J, Lim Y, Lee K, Byun HY, Yim D. Use of a Target‐Mediated Drug Disposition Model to Predict the Human Pharmacokinetics and Target Occupancy of
GC
1118, an Anti‐epidermal Growth Factor Receptor Antibody. Basic Clin Pharmacol Toxicol 2016; 120:243-249. [DOI: 10.1111/bcpt.12675] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Wan‐Su Park
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| | - Seunghoon Han
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| | - Jongtae Lee
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| | - Taegon Hong
- Department of Clinical Pharmacology Severance Hospital, Yonsei University College of Medicine Seoul Korea
| | - Jonghwa Won
- MOGAM Biotechnology Institute Yongin Gyeonggi‐do Korea
| | - Yangmi Lim
- MOGAM Biotechnology Institute Yongin Gyeonggi‐do Korea
| | - Kyuhyun Lee
- MOGAM Biotechnology Institute Yongin Gyeonggi‐do Korea
| | | | - Dong‐Seok Yim
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| |
Collapse
|
27
|
Zheng B, Yu X, Greth W, Robbie GJ. Population pharmacokinetic analysis of sifalimumab from a clinical phase IIb trial in systemic lupus erythematosus patients. Br J Clin Pharmacol 2016; 81:918-28. [PMID: 26659791 PMCID: PMC4834601 DOI: 10.1111/bcp.12864] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/23/2015] [Accepted: 12/02/2015] [Indexed: 01/05/2023] Open
Abstract
AIMS Sifalimumab, a human immunoglobulin (Ig) G1 monoclonal antibody against INF-alpha, is being studied as a treatment for systemic lupus erythematosus (SLE). This analysis characterized population pharmacokinetics (PK) of sifalimumab following repeat fixed dose and evaluated the utility of fixed dosing vs. body weight normalized dosing in SLE patients. METHODS PK data were collected in a phase IIb study where 298 patients received multiple intravenous doses (200-1200 mg) of sifalimumab every 4 weeks for 52 weeks. A population pharmacokinetic model was developed using 3961 quantifiable serum concentrations and the impact of patient demographics, clinical indices and biomarkers on pharmacokinetic parameters was evaluated. The appropriateness of the final model was evaluated using visual predictive check and bootstrap. RESULTS A two compartment model with first order elimination adequately described sifalimumab serum PK. The estimated typical clearance (CL) and central volume of distribution (V1 ) were 184 ml day(-1) and 2.82 l with 24% and 16% between-subject variability (BSV), respectively. Body weight, dose, 21 INF gene signature baseline and concomitant steroid use were identified as statistically significant covariates for CL and V1 and accounted for <10% of PK variability in the final model. Typical values and BSV of PK parameters from the current analysis with fixed dosing were similar to previous population PK results with body weight normalized dosing. CONCLUSIONS The transition from body weight normalized dosing to fixed dosing did not impact sifalimumab PK. These findings support the use of fixed dosing for sifalimumab in future clinical studies evaluating it as a potential treatment for SLE.
Collapse
Affiliation(s)
- Bo Zheng
- Clinical Pharmacology and DMPK (CPD)MedImmune, LLC, One MedImmune WayGaithersburgMaryland20878
| | - Xiang‐Qing Yu
- Clinical Pharmacology and DMPK (CPD)MedImmune, LLC, One MedImmune WayGaithersburgMaryland20878
| | - Warren Greth
- Clinical DevelopmentMedImmune, LLC, One MedImmune WayGaithersburgMaryland20878USA
| | - Gabriel J. Robbie
- Clinical Pharmacology and DMPK (CPD)MedImmune, LLC, One MedImmune WayGaithersburgMaryland20878
| |
Collapse
|
28
|
Noh H, Park MS, Kim SH, Oh SJ, Zang DY, Park HL, Cho DJ, Kim DW, Lee JI. Optimization of radotinib doses for the treatment of Asian patients with chronic myelogenous leukemia based on dose-response relationship analyses. Leuk Lymphoma 2015; 57:1856-64. [DOI: 10.3109/10428194.2015.1113278] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
29
|
Buil-Bruna N, López-Picazo JM, Martín-Algarra S, Trocóniz IF. Bringing Model-Based Prediction to Oncology Clinical Practice: A Review of Pharmacometrics Principles and Applications. Oncologist 2015; 21:220-32. [PMID: 26668254 DOI: 10.1634/theoncologist.2015-0322] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 11/17/2022] Open
Abstract
UNLABELLED Despite much investment and progress, oncology is still an area with significant unmet medical needs, with new therapies and more effective use of current therapies needed. The emergent field of pharmacometrics combines principles from pharmacology (pharmacokinetics [PK] and pharmacodynamics [PD]), statistics, and computational modeling to support drug development and optimize the use of already marketed drugs. Although it has gained a role within drug development, its use in clinical practice remains scarce. The aim of the present study was to review the principal pharmacometric concepts and provide some examples of its use in oncology. Integrated population PK/PD/disease progression models as part of the pharmacometrics platform provide a powerful tool to predict outcomes so that the right dose can be given to the right patient to maximize drug efficacy and reduce drug toxicity. Population models often can be developed with routinely collected medical record data; therefore, we encourage the application of such models in the clinical setting by generating close collaborations between physicians and pharmacometricians. IMPLICATIONS FOR PRACTICE The present review details how the emerging field of pharmacometrics can integrate medical record data with predictive pharmacological and statistical models of drug response to optimize and individualize therapies. In order to make this routine practice in the clinic, greater awareness of the potential benefits of the field is required among clinicians, together with closer collaboration between pharmacometricians and clinicians to ensure the requisite data are collected in a suitable format for pharmacometrics analysis.
Collapse
Affiliation(s)
- Núria Buil-Bruna
- Pharmacometrics and Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - José-María López-Picazo
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Salvador Martín-Algarra
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Iñaki F Trocóniz
- Pharmacometrics and Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
30
|
Lo L, Patel D, Townsend AR, Price TJ. Pharmacokinetic and pharmacodynamic evaluation of panitumumab in the treatment of colorectal cancer. Expert Opin Drug Metab Toxicol 2015; 11:1907-24. [PMID: 26572750 DOI: 10.1517/17425255.2015.1112787] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Integration of targeted therapy and additional chemotherapy options has improved median overall survival (OS) in patients with unresectable metastatic colorectal cancer (mCRC). Cetuximab and panitumumab are examples of targeted therapies, specifically against the epidermal growth factor receptor (EGFR). This review focuses on Panitumumab, a fully human IgG2 monoclonal antibody, which inhibits key oncogenic downstream cell signalling pathways. Panitumumab and cetuximab have improved tumour response rate, progression-free survival, and OS in mCRC patients in whom the RAS (Rat Sarcoma) gene is of Wild Type (WT) status. AREAS COVERED The EGFR signalling pathway and preclinical, Phase I and Phase II clinical studies on the pharmacokinetic, pharmacodynamic and safety evaluation of panitumumab are presented. Phase III studies utilising panitumumab in the first, second and third line setting in mCRC are also described. EXPERT OPINION Panitumumab exhibits excellent pharmacokinetics and pharmacodynamics by way of uncomplicated dosing, non-existent drug interactions, minimal infusion reactions and manageable side effects, making it a suitable target for combination treatments. However, innate and acquired resistances are still obstacles. To overcome this, experimented strategies are ongoing, particularly in patients with Her-2 and BRAF gene alterations. Novel biomarkers to improve patient selection and second-generation targeted antibodies are in development.
Collapse
Affiliation(s)
- Louisa Lo
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia
| | - Dainik Patel
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia
| | - Amanda R Townsend
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia.,b School of Medicine , University of Adelaide , Adelaide , 5000 , SA , Australia
| | - Timothy J Price
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia.,b School of Medicine , University of Adelaide , Adelaide , 5000 , SA , Australia
| |
Collapse
|
31
|
Rosario M, Dirks NL, Gastonguay MR, Fasanmade AA, Wyant T, Parikh A, Sandborn WJ, Feagan BG, Reinisch W, Fox I. Population pharmacokinetics-pharmacodynamics of vedolizumab in patients with ulcerative colitis and Crohn's disease. Aliment Pharmacol Ther 2015; 42:188-202. [PMID: 25996351 PMCID: PMC5032981 DOI: 10.1111/apt.13243] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/12/2015] [Accepted: 04/24/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Vedolizumab, an anti-α(4)β(7) integrin monoclonal antibody (mAb), is indicated for treating patients with moderately to severely active ulcerative colitis (UC) and Crohn's disease (CD). As higher therapeutic mAb concentrations have been associated with greater efficacy in inflammatory bowel disease, understanding determinants of vedolizumab clearance may help to optimise dosing. AIMS To characterise vedolizumab pharmacokinetics in patients with UC and CD, to identify clinically relevant determinants of vedolizumab clearance, and to describe the pharmacokinetic-pharmacodynamic relationship using population modelling. METHODS Data from a phase 1 healthy volunteer study, a phase 2 UC study, and 3 phase 3 UC/CD studies were included. Population pharmacokinetic analysis for repeated measures was conducted using nonlinear mixed effects modelling. Results from the base model, developed using extensive phase 1 and 2 data, were used to develop the full covariate model, which was fit to sparse phase 3 data. RESULTS Vedolizumab pharmacokinetics was described by a 2-compartment model with parallel linear and nonlinear elimination. Using reference covariate values, linear elimination half-life of vedolizumab was 25.5 days; linear clearance (CL(L)) was 0.159 L/day for UC and 0.155 L/day for CD; central compartment volume of distribution (V(c)) was 3.19 L; and peripheral compartment volume of distribution was 1.66 L. Interindividual variabilities (%CV) were 35% for CLL and 19% for V(c); residual variance was 24%. Only extreme albumin and body weight values were identified as potential clinically important predictors of CL(L). CONCLUSIONS Population pharmacokinetic parameters were similar in patients with moderately to severely active UC and CD. This analysis supports use of vedolizumab fixed dosing in these patients. Clinicaltrials.gov Identifiers: NCT01177228; NCT00783718 (GEMINI 1); NCT00783692 (GEMINI 2); NCT01224171 (GEMINI 3).
Collapse
Affiliation(s)
- M. Rosario
- Takeda Pharmaceuticals International Co.CambridgeMAUSA
| | | | | | | | - T. Wyant
- Takeda Pharmaceuticals International Co.CambridgeMAUSA
| | - A. Parikh
- Takeda Pharmaceuticals International, Inc.DeerfieldILUSA
| | - W. J. Sandborn
- Division of GastroenterologyUniversity of California San DiegoLa JollaCAUSA
| | - B. G. Feagan
- Robarts Research InstituteUniversity of Western OntarioLondonONCanada
| | - W. Reinisch
- Department Internal Medicine IIIMedical University of ViennaViennaAustria,Department of Internal MedicineMcMaster UniversityHamiltonONCanada
| | - I. Fox
- Takeda Pharmaceuticals International Co.CambridgeMAUSA
| |
Collapse
|
32
|
Rodríguez-Vera L, Ramos-Suzarte M, Fernández-Sánchez E, Soriano JL, Guitart CP, Hernández GC, Jacobo-Cabral CO, de Castro Suárez N, Codina HC. Semimechanistic model to characterize nonlinear pharmacokinetics of nimotuzumab in patients with advanced breast cancer. J Clin Pharmacol 2015; 55:888-98. [PMID: 25760761 DOI: 10.1002/jcph.496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study aimed (1) to develop a semimechanistic pharmacokinetic (PK) model for nimotuzumab in patients with advanced breast cancer and (2) to identify demographic, biochemical, and clinical predictive factors of the PK variability. Data from a phase 1 study were analyzed using the nonlinear mixed-effects approach (NONMEM). A target-mediated disposition model that included 2 open PK compartments, the monoclonal antibody (mAb)-target binding, and target and mAb-target complex turnovers best described the linear and nonlinear PK. Covariates had no influence on the PK parameters. The final parameter estimates were 19.93 L (steady-state volume), 0.0045-0.0172 L/h (range of total clearance values), 6.96 μg/mL (steady-state binding constant), 5.50 h(-1) (target degradation rate constant), 1.43 (μg/mL) · h(-1) (complex formation rate), and 0.148 h(-1) (complex internalization rate constant). The model described the effect of the mAb-target binding, and target and mAb-target complex turnovers on nimotuzumab PK. Simulations showed that doses above 200 mg maintained the 50% target occupancy during all of the treatment. This model can be very useful for knowing the dosing schedules required for efficacy and supports further investigation of the pharmacokinetic/pharmacodynamic relationships of nimotuzumab to improve its therapeutic use.
Collapse
Affiliation(s)
- Leyanis Rodríguez-Vera
- Laboratory of Pharmacokinetic, Department of Pharmacology & Clinical Pharmacy, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | | | - Eduardo Fernández-Sánchez
- Center for Biological Evaluation and Research, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | | | - Concepción Peraire Guitart
- Pharmacy and Pharmaceutical Technology Department, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | | | | | - Niurys de Castro Suárez
- Laboratory of Pharmacokinetic, Department of Pharmacology & Clinical Pharmacy, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | - Helena Colom Codina
- Pharmacy and Pharmaceutical Technology Department, School of Pharmacy, University of Barcelona, Barcelona, Spain
| |
Collapse
|
33
|
Model-based assessment of dosing strategies in children for monoclonal antibodies exhibiting target-mediated drug disposition. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e138. [PMID: 25271939 PMCID: PMC4474168 DOI: 10.1038/psp.2014.38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/18/2014] [Indexed: 01/04/2023]
Abstract
Body weight/body surface area–based and/or tiered fixed dosing strategies are widely utilized for monoclonal antibodies with linear clearance to scale adult clinical doses to children. However, there is limited knowledge on whether or not body weight–based dosing strategies also yield comparable dose-concentration-response relationships in adults and children for monoclonal antibodies that exhibit target-mediated drug disposition. Our findings indicate that it is important to interpret pharmacokinetics information in a pharmacokinetics/pharmacodynamics context as similar systemic drug exposure in adults and children may not be reflective of the corresponding target occupancy. They further indicate that BW-based dosing is superior to fixed dosing for the same target concentration, whereas the opposite holds true for the same target amount in adults and children. Michaelis-Menten approximations yielded similar profiles compared to the full target-mediated drug disposition model for all simulation scenarios and may be used to guide the selection of appropriate dosing regimens in children.
Collapse
|
34
|
Guo Y, Shi M, Yang A, Feng J, Zhu X, Choi YJ, Hu G, Pan J, Hu C, Luo R, Zhang Y, Zhou L, Cheng Y, Lüpfert C, Cai J, Shi Y. Platinum-based chemotherapy plus cetuximab first-line for Asian patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck: Results of an open-label, single-arm, multicenter trial. Head Neck 2014; 37:1081-7. [PMID: 24710768 DOI: 10.1002/hed.23707] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 01/28/2014] [Accepted: 04/04/2014] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The purpose of this study was to assess the efficacy, safety, and pharmacokinetics of cisplatin-based chemotherapy plus cetuximab as first-line treatment in Chinese and Korean patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck (SCCHN). METHODS Patients (n = 68) received cetuximab weekly plus 3-week cycles of cisplatin/5-fluorouracil (5-FU) chemotherapy for up to 6 cycles. The primary endpoint was overall response rate. RESULTS The overall response rate was 55.9%, including 2 complete responses (CRs). Median overall survival (OS) was 12.6 months and median progression-free survival (PFS) was 6.6 months. Grade 3/4 adverse events (AEs) were reported in 41 (60.3%) patients. The safety profile was in line with previous clinical experience. The pharmacokinetic profile was in line with that observed with cetuximab in white and Japanese patients. CONCLUSION The efficacy, safety, and pharmacokinetic findings from this study support the use of first-line platinum-based chemotherapy plus cetuximab in Chinese and Korean patients with recurrent and/or metastatic SCCHN (ClinicalTrials.gov NCT01177956).
Collapse
Affiliation(s)
- Ye Guo
- Medical Oncology Department, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mei Shi
- Radiation Oncology Department, Xijing Hospital - The First Affiliated Hospital of the Fourth Military Medical University, Shanxi Province, China
| | - Ankui Yang
- Head and Neck Department, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jifeng Feng
- Medical Oncology Department, Jiangsu Cancer Hospital, Nanjing, Jiangsu Province, China
| | - Xiaodong Zhu
- Radiotherapy Department, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Province, China
| | - Young-Jin Choi
- Medical Oncology Department, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Guoqin Hu
- Cancer Department, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jianji Pan
- Department of Radiation Oncology, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Chunhong Hu
- Oncology Department, The Second Xiangya Hospital of Central South University, Changsha, Hu'nan Province, China
| | - Rongcheng Luo
- Cancer Medical Department, Nanfang Hospital, Guangzhou, China
| | - Yiping Zhang
- Chemotherapy Center, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Liang Zhou
- Head and Neck Surgery Department, Eye and Ear, Nose, and Throat Hospital of Fudan University, Xuhui District, Shanghai, China
| | - Ying Cheng
- Medical Oncology Department, Jilin Cancer Hospital, Changchun, Jilin, China
| | | | - Junliang Cai
- Merck Serono (Beijing) Pharmaceutical R&D Company, The Center, Shanghai, China
| | - Yuankai Shi
- Department of Medical Oncology, Cancer Institute/Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Chaoyang District, Beijing, China
| |
Collapse
|
35
|
Ingargiola M, Runge R, Heldt JM, Freudenberg R, Steinbach J, Cordes N, Baumann M, Kotzerke J, Brockhoff G, Kunz-Schughart LA. Potential of a Cetuximab-based radioimmunotherapy combined with external irradiation manifests in a 3-D cell assay. Int J Cancer 2014; 135:968-80. [PMID: 24615356 DOI: 10.1002/ijc.28735] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/04/2013] [Accepted: 01/08/2014] [Indexed: 12/22/2022]
Abstract
Targeting epidermal growth factor receptor (EGFR)-overexpressing tumors with radiolabeled anti-EGFR antibodies is a promising strategy for combination with external radiotherapy. In this study, we evaluated the potential of external plus internal irradiation by [(90) Y]Y-CHX-A″-DTPA-C225 (Y-90-C225) in a 3-D environment using FaDu and SAS head and neck squamous cell carcinoma (HNSCC) spheroid models and clinically relevant endpoints such as spheroid control probability (SCP) and spheroid control dose 50% (SCD50 , external irradiation dose inducing 50% loss of spheroid regrowth). Spheroids were cultured using a standardized platform. Therapy response after treatment with C225, CHX-A"-DTPA-C225 (DTPA-C225), [(90) Y]Y-CHX-A"-DTPA (Y-90-DTPA) and Y-90-C225 alone or in combination with X-ray was evaluated by long-term monitoring (60 days) of spheroid integrity and volume growth. Penetration kinetics into spheroids and EGFR binding capacities on spheroid cells were identical for unconjugated C225 and Y-90-C225. Spheroid-associated radioactivity upon exposure to the antibody-free control conjugate Y-90-DTPA was negligible. Determination of the SCD50 demonstrated higher intrinsic radiosensitivity of FaDu as compared with SAS spheroids. Treatment with unconjugated C225 alone did not affect spheroid growth and cell viability. Also, C225 treatment after external irradiation showed no additive effect. However, the combination of external irradiation with Y-90-C225 (1 µg/ml, 24 hr) resulted in a considerable benefit as reflected by a pronounced reduction of the SCD50 from 16 Gy to 9 Gy for SAS spheroids and a complete loss of regrowth for FaDu spheroids due to the pronounced accumulation of internal dose caused by the continuous exposure to cell-bound radionuclide upon Y-90-C225-EGFR interaction.
Collapse
Affiliation(s)
- M Ingargiola
- OncoRay-National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany; Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fronton L, Pilari S, Huisinga W. Monoclonal antibody disposition: a simplified PBPK model and its implications for the derivation and interpretation of classical compartment models. J Pharmacokinet Pharmacodyn 2014; 41:87-107. [PMID: 24493102 DOI: 10.1007/s10928-014-9349-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 01/15/2014] [Indexed: 11/24/2022]
Abstract
The structure, interpretation and parameterization of classical compartment models as well as physiologically-based pharmacokinetic (PBPK) models for monoclonal antibody (mAb) disposition are very diverse, with no apparent consensus. In addition, there is a remarkable discrepancy between the simplicity of experimental plasma and tissue profiles and the complexity of published PBPK models. We present a simplified PBPK model based on an extravasation rate-limited tissue model with elimination potentially occurring from various tissues and plasma. Based on model reduction (lumping), we derive several classical compartment model structures that are consistent with the simplified PBPK model and experimental data. We show that a common interpretation of classical two-compartment models for mAb disposition-identifying the central compartment with the total plasma volume and the peripheral compartment with the interstitial space (or part of it)-is not consistent with current knowledge. Results are illustrated for the monoclonal antibodies 7E3 and T84.66 in mice.
Collapse
Affiliation(s)
- Ludivine Fronton
- Institute of Biochemistry and Biology, Universität Potsdam, Potsdam, Germany
| | | | | |
Collapse
|
37
|
Narwal R, Roskos LK, Robbie GJ. Population pharmacokinetics of sifalimumab, an investigational anti-interferon-α monoclonal antibody, in systemic lupus erythematosus. Clin Pharmacokinet 2013; 52:1017-27. [PMID: 23754736 PMCID: PMC3824374 DOI: 10.1007/s40262-013-0085-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Sifalimumab is a fully human immunoglobulin G1κ monoclonal antibody that binds to and neutralizes a majority of the subtypes of human interferon-α. Sifalimumab is being evaluated as a treatment for systemic lupus erythematosus (SLE). The primary objectives of this analysis were (a) to develop a population pharmacokinetic model for sifalimumab in SLE; (b) to identify and quantitate the impact of patient/disease characteristics on pharmacokinetic variability; and (c) to evaluate fixed versus body weight (WT)-based dosing regimens. METHODS Sifalimumab serum concentration-time data were collected from a phase Ib study (MI-CP152) designed to evaluate the safety and tolerability of sifalimumab in adult patients with SLE. Sifalimumab was administered every 14 days as a 30- to 60-minute intravenous infusion with escalating doses of 0.3, 1.0, 3.0, and 10 mg/kg and serum concentrations were collected over 350 days. A total of 120 patients provided evaluable pharmacokinetic data with a total of 2,370 serum concentrations. Sifalimumab serum concentrations were determined using a validated colorimetric enzyme-linked immunosorbent assay (ELISA) with a lower limit of quantitation of 1.25 μg/mL. Population pharmacokinetic modeling of sifalimumab was performed using a non-linear mixed effects modeling approach with NONMEM VII software. Impact of patient demographics, clinical indices, and biomarkers on pharmacokinetic parameters were explored using a stepwise forward selection and backward elimination approach. The appropriateness of the final model was tested using visual predictive check (VPC). The impact of body WT-based and fixed dosing of sifalimumab was evaluated using a simulation approach. The final population model was utilized for phase IIb dosing projections. RESULTS Sifalimumab pharmacokinetics were best described using a two-compartment linear model with first order elimination. Following intravenous dosing, the typical clearance (CL) and central volume of distribution (V 1) were estimated to be 176 mL/day and 2.9 L, respectively. The estimates (coefficient of variation) of between-subject variability for CL and V 1 were 28 and 31 %, respectively. Patient baseline body WT, interferon gene signature from 21 genes, steroid use, and sifalimumab dose were identified as significant covariates for CL, whereas only baseline body WT was a significant covariate for V 1 and peripheral volume of distribution (V 2). Although the above-mentioned covariates were statistically significant, they did not explain variability in pharmacokinetic parameters to any relevant extent (<7 %). Thus, no dosing adjustments are necessary. VPC confirmed good predictability of the final population pharmacokinetic model. Simulation results demonstrate that both fixed and body WT-based dosing regimens yield similar median steady state concentrations and overall variability. Fixed sifalimumab doses of 200, 600, and 1,200 mg monthly (with a loading dose at Day 14) were selected for a phase IIb clinical trial. CONCLUSION A two-compartment population pharmacokinetic model adequately described sifalimumab pharmacokinetics. The estimated typical pharmacokinetic parameters were similar to other monoclonal antibodies without target mediated elimination. Although the population pharmacokinetic analysis identified some statistically significant covariates, they explained <7 % between-subject variability in pharmacokinetic parameters indicating that these covariates are not clinically relevant. The population pharmacokinetic analysis also demonstrated the feasibility of switching to fixed doses in phase IIb clinical trials of sifalimumab.
Collapse
Affiliation(s)
- Rajesh Narwal
- Clinical Pharmacology and DMPK (CPD), MedImmune, LLC One MedImmune Way, Gaithersburg, MD, 20878, USA,
| | | | | |
Collapse
|
38
|
Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 2013; 52:83-124. [PMID: 23299465 DOI: 10.1007/s40262-012-0027-4] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Development of monoclonal antibodies (mAbs) and their functional derivatives represents a growing segment of the development pipeline in the pharmaceutical industry. More than 25 mAbs and derivatives have been approved for a variety of therapeutic applications. In addition, around 500 mAbs and derivatives are currently in different stages of development. mAbs are considered to be large molecule therapeutics (in general, they are 2-3 orders of magnitude larger than small chemical molecule therapeutics), but they are not just big chemicals. These compounds demonstrate much more complex pharmacokinetic and pharmacodynamic behaviour than small molecules. Because of their large size and relatively poor membrane permeability and instability in the conditions of the gastrointestinal tract, parenteral administration is the most usual route of administration. The rate and extent of mAb distribution is very slow and depends on extravasation in tissue, distribution within the particular tissue, and degradation. Elimination primarily happens via catabolism to peptides and amino acids. Although not definitive, work has been published to define the human tissues mainly involved in the elimination of mAbs, and it seems that many cells throughout the body are involved. mAbs can be targeted against many soluble or membrane-bound targets, thus these compounds may act by a variety of mechanisms to achieve their pharmacological effect. mAbs targeting soluble antigen generally exhibit linear elimination, whereas those targeting membrane-bound antigen often exhibit non-linear elimination, mainly due to target-mediated drug disposition (TMDD). The high-affinity interaction of mAbs and their derivatives with the pharmacological target can often result in non-linear pharmacokinetics. Because of species differences (particularly due to differences in target affinity and abundance) in the pharmacokinetics and pharmacodynamics of mAbs, pharmacokinetic/pharmacodynamic modelling of mAbs has been used routinely to expedite the development of mAbs and their derivatives and has been utilized to help in the selection of appropriate dose regimens. Although modelling approaches have helped to explain variability in both pharmacokinetic and pharmacodynamic properties of these drugs, there is a clear need for more complex models to improve understanding of pharmacokinetic processes and pharmacodynamic interactions of mAbs with the immune system. There are different approaches applied to physiologically based pharmacokinetic (PBPK) modelling of mAbs and important differences between the models developed. Some key additional features that need to be accounted for in PBPK models of mAbs are neonatal Fc receptor (FcRn; an important salvage mechanism for antibodies) binding, TMDD and lymph flow. Several models have been described incorporating some or all of these features and the use of PBPK models are expected to expand over the next few years.
Collapse
|
39
|
Abstract
Head and neck cancer is the sixth most common cancer worldwide. At present, globally about 650,000 new cases of squamous cell carcinoma of the head and neck (SCCHN) are diagnosed each year. The epidermal growth factor receptor (EGFR) is almost invariably expressed in SCCHN. Overexpression of the EGFR is a strong and independent unfavorable prognostic factor in SCCHN. Cetuximab is a chimeric monoclonal antibody, which binds with high affinity to the extracellular domain of the human EGFR, blocking ligand binding, resulting in inhibition of the receptor function. It also targets cytotoxic immune effector cells towards EGFR-expressing tumor cells (antibody dependent cell-mediated cytotoxicity). The addition of cetuximab to radiotherapy (RT) improves locoregional control and survival when compared to RT alone. The addition of cetuximab to platinum-based chemoradiation (CRT) is feasible but does not lead to an improved outcome. Cetuximab plus RT has never been compared prospectively to CRT, which therefore remains the standard treatment for patients with locoregionally advanced SCCHN for whom surgery is not considered the optimal treatment, provided they can tolerate CRT. The addition of cetuximab to platinum-based chemotherapy prolongs survival in patients with recurrent or metastatic SCCHN. The combination of a platinum-based regimen and cetuximab should be considered as the standard first line regimen for patients who can tolerate this treatment.
Collapse
Affiliation(s)
- Pol Specenier
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Jan B Vermorken
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
40
|
Gao B, Yeap S, Clements A, Balakrishnar B, Wong M, Gurney H. Evidence for Therapeutic Drug Monitoring of Targeted Anticancer Therapies. J Clin Oncol 2012; 30:4017-25. [DOI: 10.1200/jco.2012.43.5362] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Therapeutic drug monitoring (TDM) provides valuable guidance for dose adjustment of antibiotics, immunosuppressives, antiepileptics, and other drugs, but its use for traditional anticancer therapies has been limited. Perhaps the most important obstacle is the impractical requirement of multiple blood samples to adequately define systemic exposure of drugs that have a short elimination half-life and are given by intermittent intravenous injections. However, the newer targeted anticancer therapies have different pharmacokinetic (PK) and dosing characteristics compared with traditional cytotoxic drugs, making it possible to estimate the steady-state drug exposure with a single trough-level measurement. Recent evidence indicates that certain PK parameters, including trough levels, are correlated with clinical outcomes for many of these agents, including imatinib, sunitinib, rituximab, and cetuximab. Although the current evidence is insufficient to mandate TDM in routine practice, a concerted investigation should be encouraged to determine whether the steady-state trough measurements of targeted agents will have a practical place in the clinical care of patients with cancer.
Collapse
Affiliation(s)
- Bo Gao
- Bo Gao, Arthur Clements, Bavanthi Balakrishnar, Mark Wong, and Howard Gurney, Westmead Hospital, Westmead; Bo Gao and Howard Gurney, University of Sydney, Sydney, New South Wales, Australia; and Shang Yeap, Johns Hopkins Hospital,Singapore
| | - Shang Yeap
- Bo Gao, Arthur Clements, Bavanthi Balakrishnar, Mark Wong, and Howard Gurney, Westmead Hospital, Westmead; Bo Gao and Howard Gurney, University of Sydney, Sydney, New South Wales, Australia; and Shang Yeap, Johns Hopkins Hospital,Singapore
| | - Arthur Clements
- Bo Gao, Arthur Clements, Bavanthi Balakrishnar, Mark Wong, and Howard Gurney, Westmead Hospital, Westmead; Bo Gao and Howard Gurney, University of Sydney, Sydney, New South Wales, Australia; and Shang Yeap, Johns Hopkins Hospital,Singapore
| | - Bavanthi Balakrishnar
- Bo Gao, Arthur Clements, Bavanthi Balakrishnar, Mark Wong, and Howard Gurney, Westmead Hospital, Westmead; Bo Gao and Howard Gurney, University of Sydney, Sydney, New South Wales, Australia; and Shang Yeap, Johns Hopkins Hospital,Singapore
| | - Mark Wong
- Bo Gao, Arthur Clements, Bavanthi Balakrishnar, Mark Wong, and Howard Gurney, Westmead Hospital, Westmead; Bo Gao and Howard Gurney, University of Sydney, Sydney, New South Wales, Australia; and Shang Yeap, Johns Hopkins Hospital,Singapore
| | - Howard Gurney
- Bo Gao, Arthur Clements, Bavanthi Balakrishnar, Mark Wong, and Howard Gurney, Westmead Hospital, Westmead; Bo Gao and Howard Gurney, University of Sydney, Sydney, New South Wales, Australia; and Shang Yeap, Johns Hopkins Hospital,Singapore
| |
Collapse
|
41
|
Johnson DE. Biotherapeutic first-in-human dose selection: making use of preclinical markers. Expert Rev Clin Pharmacol 2012; 3:231-42. [PMID: 22111569 DOI: 10.1586/ecp.10.5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
First-in-human dose-selection criteria for biotherapeutics are changing, primarily based on severe adverse events in a single monoclonal antibody trial in healthy volunteers. Spurred by new EMA guidance, the minimum anticipated biological-effect level (MABEL) for estimating a starting human dose from exposure-response preclinical data have been introduced and should help to create long overdue target mechanism-based models focused on exposure-response relationships. Even though clarity of its application is still developing, this has the potential to become the model for most biotherapeutics in the future. However, maximizing benefit from MABEL will require increased efforts to define and create assays for relevant biomarkers of biological activity and safety as pharmacodynamic end points. Currently, this has not been realized sufficiently to make the model applicable to a majority of biotherapeutics; however, this review suggests how it can be applied universally with monoclonal antibodies.
Collapse
Affiliation(s)
- Dale E Johnson
- Emiliem, Inc., 6027 Christie Avenue, Emeryville, CA 94608, USA and University of California, Berkeley, Morgan Hall, Berkeley, CA 94720-3104 USA.
| |
Collapse
|
42
|
Bai S, Jorga K, Xin Y, Jin D, Zheng Y, Damico-Beyer LA, Gupta M, Tang M, Allison DE, Lu D, Zhang Y, Joshi A, Dresser MJ. A guide to rational dosing of monoclonal antibodies. Clin Pharmacokinet 2012; 51:119-35. [PMID: 22257150 DOI: 10.2165/11596370-000000000-00000] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE Dosing of therapeutic monoclonal antibodies (mAbs) is often based on body size, with the perception that body size-based dosing would reduce inter-subject variability in drug exposure. However, most mAbs are target specific with a relatively large therapeutic window and generally a small contribution of body size to pharmacokinetic variability. Therefore, the dosing paradigm for mAbs should be assessed in the context of these unique characteristics. The objective of this study was to review the current dosing strategy and to provide a scientific rationale for dosing of mAbs using a modelling and simulation approach. METHODS In this analysis, the body weight-based or body weight-independent (fixed) dosing regimens for mAbs were systematically evaluated. A generic two-compartment first-order elimination model was developed. Individual or population pharmacokinetic profiles were simulated as a function of the body weight effects on clearance (θ(BW_CL)) and on the central volume of distribution (θ(BW_V1)). The variability in exposure (the area under the serum concentration-time curve [AUC], trough serum concentration [C(min)] and peak serum concentration [C(max)]) was compared between body weight-based dosing and fixed dosing in the entire population. The deviation of exposure for light and heavy subjects from median body weight subjects was also measured. The simulation results were then evaluated with clinical pharmacokinetic characteristics of various mAbs that were given either by body weight-based dosing or by fixed dosing in the case study. RESULTS Results from this analysis demonstrated that exposure variability was dependent on the magnitude of the body weight effect on pharmacokinetics. In contrast to the conventional assumption, body weight-based dosing does not always offer advantages over fixed dosing in reducing exposure variability. In general, when the exponential functions of θ(BW_CL) and θ(BW_V1) in the population pharmacokinetic model are <0.5, fixed dosing results in less variability and less deviation than body weight-based dosing; when both θ(BW_CL) and θ(BW_V1) are >0.5, body weight-based dosing results in less variability and less deviation than fixed dosing. In the scenarios when either θ(BW_CL) or θ(BW_V1) is >0.5, the impact on exposure variability is different for each exposure measure. The case study demonstrated that most mAbs had little effect or a moderate body weight effect (θ(BW_CL) and θ(BW_V1) <0.5 or ∼0.5). The difference of variability in exposure between body weight-based and fixed dosing is generally less than 20% and the percentages of deviation for light and heavy subpopulations are less than 40%. CONCLUSIONS The analysis provided insights into the conditions under which either fixed or body weight-based dosing would be superior in reducing pharmacokinetic variability and exposure differences between light and heavy subjects across the population. The pharmacokinetic variability introduced by either dosing regimen is moderate relative to the variability generally observed in pharmacodynamics, efficacy and safety. Therefore, mAb dosing can be flexible. Given many practical advantages, fixed dosing is recommended to be the first option in first-in-human studies with mAbs. The dosing strategy in later stages of clinical development could then be determined based on combined knowledge of the body weight effect on pharmacokinetics, safety and efficacy from the early clinical trials.
Collapse
Affiliation(s)
- Shuang Bai
- Genentech Inc., South San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Krippendorff BF, Oyarzún DA, Huisinga W. Predicting the F(ab)-mediated effect of monoclonal antibodies in vivo by combining cell-level kinetic and pharmacokinetic modelling. J Pharmacokinet Pharmacodyn 2012; 39:125-39. [PMID: 22399130 PMCID: PMC3333800 DOI: 10.1007/s10928-012-9243-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 02/09/2012] [Indexed: 01/09/2023]
Abstract
Cell-level kinetic models for therapeutically relevant processes increasingly benefit the early stages of drug development. Later stages of the drug development processes, however, rely on pharmacokinetic compartment models while cell-level dynamics are typically neglected. We here present a systematic approach to integrate cell-level kinetic models and pharmacokinetic compartment models. Incorporating target dynamics into pharmacokinetic models is especially useful for the development of therapeutic antibodies because their effect and pharmacokinetics are inherently interdependent. The approach is illustrated by analysing the F(ab)-mediated inhibitory effect of therapeutic antibodies targeting the epidermal growth factor receptor. We build a multi-level model for anti-EGFR antibodies by combining a systems biology model with in vitro determined parameters and a pharmacokinetic model based on in vivo pharmacokinetic data. Using this model, we investigated in silico the impact of biochemical properties of anti-EGFR antibodies on their F(ab)-mediated inhibitory effect. The multi-level model suggests that the F(ab)-mediated inhibitory effect saturates with increasing drug-receptor affinity, thereby limiting the impact of increasing antibody affinity on improving the effect. This indicates that observed differences in the therapeutic effects of high affinity antibodies in the market and in clinical development may result mainly from Fc-mediated indirect mechanisms such as antibody-dependent cell cytotoxicity.
Collapse
Affiliation(s)
- Ben-Fillippo Krippendorff
- Pharmacology & Drug Development Group, Department of Oncology, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE UK
| | - Diego A. Oyarzún
- Centre for Synthetic Biology and Innovation, Department of Bioengineering, Imperial College London, London, SW7 2AZ UK
| | - Wilhelm Huisinga
- Institut für Mathematik, Universität Potsdam, Wissenschaftspark Golm, 14476 Potsdam, Germany
| |
Collapse
|
44
|
Azzopardi N, Lecomte T, Ternant D, Boisdron-Celle M, Piller F, Morel A, Gouilleux-Gruart V, Vignault-Desvignes C, Watier H, Gamelin E, Paintaud G. Cetuximab Pharmacokinetics Influences Progression-Free Survival of Metastatic Colorectal Cancer Patients. Clin Cancer Res 2011; 17:6329-37. [DOI: 10.1158/1078-0432.ccr-11-1081] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Specenier P, Vermorken JB. Cetuximab in the treatment of squamous cell carcinoma of the head and neck. Expert Rev Anticancer Ther 2011; 11:511-24. [PMID: 21504318 DOI: 10.1586/era.11.20] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The majority of the head and neck cancers are squamous cell carcinomas, which commonly overexpress the EGF receptor (EGFR). Cetuximab is a chimeric monoclonal antibody that binds with high affinity to the extracellular domain of EGFR, and in addition induces antibody-dependent cellular cytoxicity. In a randomized Phase III trial in patients with locoregionally advanced squamous cell carcinoma of the head and neck, the addition of cetuximab to radiotherapy prolonged the median time of locoregional control from 14.9 to 24.4 months and increased the median overall survival from 29.3 to 49.0 months. In patients with platinum-refractory recurrent and/or metastatic disease, the objective response and disease-control rates in various studies ranged from 10 to 13% and from 46 to 56%, respectively. In the EXTREME trial, the addition of cetuximab to platinum/5-fluorouracil as first-line treatment of recurrent/metastatic squamous cell carcinoma of the head and neck not only led to significant improvements in survival, response rate and disease control, but also induced a better symptom control in comparison with that observed with platinum/5-fluorouracil alone.
Collapse
Affiliation(s)
- Pol Specenier
- Department of Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium.
| | | |
Collapse
|
46
|
Kakkar T, Sung C, Gibiansky L, Vu T, Narayanan A, Lin SL, Vincent M, Banfield C, Colbert A, Hoofring S, Starcevic M, Ma P. Population PK and IgE pharmacodynamic analysis of a fully human monoclonal antibody against IL4 receptor. Pharm Res 2011; 28:2530-42. [PMID: 21604075 DOI: 10.1007/s11095-011-0481-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 05/13/2011] [Indexed: 01/13/2023]
Abstract
PURPOSE For AMG 317, a fully human monoclonal antibody to interleukin receptor IL-4Rα, we developed a population pharmacokinetic (PK) model by fitting data from four early phase clinical trials of intravenous and subcutaneous (SC) routes simultaneously, investigated important PK covariates, and explored the relationship between exposure and IgE response. METHODS Data for 294 subjects and 2183 AMG 317 plasma concentrations from three Phase 1 and 1 Phase 2 studies were analyzed by nonlinear mixed effects modeling using first-order conditional estimation with interaction. The relationship of IgE response with post hoc estimates of exposure generated from the final PK model was explored based on data from asthmatic patients. RESULTS The best structural model was a two-compartment quasi-steady-state target-mediated drug disposition model with linear and non-linear clearances. For a typical 80-kg, 40-year subject, linear clearance was 35.0 mL/hr, central and peripheral volumes of distribution were 1.78 and 5.03 L, respectively, and SC bioavailability was 24.3%. Body weight was an important covariate on linear clearance and central volume; age influenced absorption rate. A significant treatment effect was observable between the cumulative AUC and IgE response measured. CONCLUSION The population PK model adequately described AMG 317 PK from IV and SC routes over a 60-fold range of doses with two dosing strengths across multiple studies covering healthy volunteers and patients with mild to severe asthma. IgE response across a range of doses and over the sampling time points was found to be related to cumulative AMG 317 exposure.
Collapse
Affiliation(s)
- Tarundeep Kakkar
- Department of Pharmacokinetics and Metabolism, Amgen Inc., Thousand Oaks, California, 91320, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yang BB, Lum P, Chen A, Arends R, Roskos L, Smith B, Pérez Ruixo JJ. Pharmacokinetic and pharmacodynamic perspectives on the clinical drug development of panitumumab. Clin Pharmacokinet 2011; 49:729-40. [PMID: 20923247 DOI: 10.2165/11535970-000000000-00000] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Panitumumab is a recombinant, fully human IgG2 monoclonal antibody directed against the epidermal growth factor receptor (EGFR). It is indicated for use as monotherapy in the treatment of patients with EGFR-expressing metastatic colorectal cancer after disease progression with standard chemotherapy. The currently indicated dose is 6 mg/kg given every 2 weeks. Panitumumab is mainly distributed into the vascular space and exhibits nonlinear pharmacokinetics that are consistent with target-mediated drug disposition, involving saturable binding to EGFR and subsequent internalization and degradation inside the cells. Panitumumab is also cleared in a linear fashion by the reticuloendothelial system, similarly to other endogenous immunoglobulins. After single-dose administration of panitumumab as a 1-hour intravenous infusion, the area under the serum concentration-time curve increases in a greater-than-dose-proportional manner as the dose increases from 0.75 to 5 mg/kg; however, at doses above 2 mg/kg, the exposure to panitumumab increases in a dose-proportional manner. Panitumumab pharmacokinetics are not meaningfully affected by the tumour type, EGFR membrane expression, tumour KRAS mutation, sex, age, race or renal or hepatic dysfunction. In addition, irinotecan-containing and paclitaxel/carboplatin-containing chemotherapeutic regimens do not appear to affect panitumumab pharmacokinetics. The results of population pharmacokinetic analyses have shown that bodyweight is the most influential covariate on panitumumab exposure, supporting the current use of bodyweight-adjusted doses (mg/kg). The relationship between the weekly dose of panitumumab and skin rash, an on-target pharmacodynamic effect of EGFR inhibition, reaches a plateau at 2.5 mg/kg, indicating that this is the optimal weekly dose. Two less-frequent dosing regimens (6 mg/kg given every 2 weeks and 9 mg/kg given every 3 weeks) achieve steady-state serum trough concentrations similar to those achieved by 2.5 mg/kg given every week, ensuring maximal EGFR coverage. Anti-panitumumab antibody production is uncommon and does not appear to have an impact on the pharmacokinetics of panitumumab.
Collapse
Affiliation(s)
- Bing-Bing Yang
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California 91320, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Dirks NL, Meibohm B. Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet 2011; 49:633-59. [PMID: 20818831 DOI: 10.2165/11535960-000000000-00000] [Citation(s) in RCA: 366] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A growing number of population pharmacokinetic analyses of therapeutic monoclonal antibodies (mAbs) have been published in the scientific literature. The aims of this article are to summarize the findings from these studies and to relate the findings to the general pharmacokinetic and structural characteristics of therapeutic mAbs. A two-compartment model was used in the majority of the population analyses to describe the disposition of the mAb. Population estimates of the volumes of distribution in the central (V(1)) and peripheral (V(2)) compartments were typically small, with median (range) values of 3.1 (2.4-5.5) L and 2.8 (1.3-6.8) L, respectively. The estimated between-subject variability in the V(1) was usually moderate, with a median (range) coefficient of variation (CV) of 26% (12-84%). Between-subject variability in other distribution-related parameters such as the V(2) and intercompartmental clearance were often not estimated. Although the pharmacokinetic models used most frequently in the population analyses were models with linear clearance, other models with nonlinear, or parallel linear and nonlinear clearance pathways were also applied, as many therapeutic mAbs are eliminated via saturable target-mediated mechanisms. Population estimates of the maximum elimination rate (V(max)) and the mAb concentration at which elimination was at half maximum for Michaelis-Menten-type elimination pathways varied considerably among the different therapeutic mAbs. However, estimates of the total clearance (CL) of mAbs with linear clearance characteristics and of the clearance of mAbs via the linear clearance pathway (CL(L)) with parallel linear and nonlinear clearance were quite similar for the different mAbs and typically ranged from 0.2 to 0.5 L/day, which is relatively close to the estimated clearance of endogenous IgG of 0.21 L/day. The between-subject variability in the V(max), CL and CL(L) was moderate to high, with estimated CVs ranging from 15% to 65%. Measures of body size were the covariates most commonly identified as influencing the pharmacokinetics of therapeutic mAbs. In summary, many features of the population pharmacokinetics of currently used therapeutic mAbs are similar, despite differences in their pharmacological targets and studied patient populations.
Collapse
Affiliation(s)
- Nathanael L Dirks
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | |
Collapse
|
50
|
Wang C, He X, Zhou B, Li J, Li B, Qian W, Hou S, Wang H, Shi Y, Guo Y. Phase 1 study of anti-epidermal growth factor receptor monoclonal antibody in patients with solid tumors. MAbs 2011; 3:67-75. [PMID: 21051930 DOI: 10.4161/mabs.3.1.14021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In the present study, we conducted a Phase 1 study of a recombinant anti-EGFR monoclonal antibody (CMAB009) that has the same amino acid sequence as cetuximab. The purpose of this study was to evaluate the safety, pharmacokinetics, and potential benefit of CMAB009 in Chinese patients with advanced chemotherapy-resistant epithelial malignancies. In this study, 18 patients were treated with two successive treatment schedules comprising a single-dose escalation phase followed by a weekly, multiple-dose extension phase. No dose-limiting toxicity was reported during the evaluation period. CMAB009-associated toxicity was minimal, and the most commonly reported adverse events were fever, asthenia, transaminase elevation, nausea, and skin toxicities. CMAB009 exhibited a non-linear PK profile over the dose range of 100 to 400 mg/m2. In the single-dose phase, CMAB009 reached peak serum concentrations at the end of the infusion and then declined slowly with a Tl/2 of 77.15 ±13.96 h, 79.79 ±6.91 h, and 86.25 ±9.93 h after infusion of 100, 250, and 400 mg/m ( 2) based on a two compartmental model analysis. Mean Cmax increased roughly dose-proportional while AUC0-∞ showed a greater than dose-proportionate increase from 100 to 400 mg/m ( 2) . After multiple infusions, serum concentrations dropped slowly and the Tl/2 was 102.25 ± 33.54 h and 118.91 ± 29.12 h based on a two compartmental model analysis. No neutralizing anti-antibody antibodies were detectable. Two patients achieved partial remissions. The study results suggest that CMAB009 shows acceptable tolerance and primary efficacy and should be studied as a treatment in patients with advanced chemotherapy-resistant epithelial malignancies.
Collapse
Affiliation(s)
- Chong Wang
- School of Medicine and School of Pharmacy, The Center for Antibody Medicine of Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|