1
|
Wanselius M, Abrahmsén-Alami S, Hanafy BI, Mazza M, Hansson P. A microfluidic in vitro method predicting the fate of peptide drugs after subcutaneous administration. Int J Pharm 2024; 667:124849. [PMID: 39454976 DOI: 10.1016/j.ijpharm.2024.124849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
For many biopharmaceuticals, subcutaneous (sc) administration is the only viable route. However, there is no in vitro method available accurately predicting the absorption profiles of subcutaneously injected pharmaceuticals. In this work, we show that a recently developed microfluidics method for interaction studies (MIS) has the potential to be useful in this respect. The method utilises the responsiveness of polyelectrolyte microgel networks to oppositely charged molecules as a means to monitor the interaction between peptides and hyaluronic acid (HA), a major constituent of the subcutaneous extracellular matrix. We use the method to determine parameters describing the strength of interaction between peptide and HA as well as the peptide's aggregation tendency and transport properties in HA networks. The results from MIS studies of the peptide drugs exenatide, pramlintide, vancomycin, polymyxin B, lanreotide, MEDI7219 and AZD2820 are compared with results from measurements with the commercially available SCISSOR system and in vivo absorption and bioavailability data from the literature. We show that both MIS and SCISSOR reveal differences in the peptides' diffusivity and tendency to aggregate in the presence of HA. We show that MIS is particularly good at discriminating between peptides forming aggregates stabilised by non-electrostatic forces in the presence of HA, and peptides forming complexes stabilised by electrostatic interactions with HA. The method provides two parameters that can be used to quantify the peptides' aggregation tendency, the one describing the peptide packing density in complexes with HA and the other the apparent diffusivity upon release in a medium of physiological ionic strength and pH. The order of the peptides when ranked by increasing binding strength at pH 7.4 determined with MIS is shown to be in agreement with the order when ranked by the apparent 1st order absorption rate constant (ka) after sc administration in humans: lanreotide (Autogel) < exenatide (IRF) < AZD2820 < pramlintide < lanreotide (IRF) (IRF: Immediate release formulation). A correlation is found between the 1st order release rate constant determined with SCISSOR and ka for lanreotide (Autogel), exenatide and AZD2820. A mechanism relating the magnitude of ka to the peptides' charge is proposed.
Collapse
Affiliation(s)
- Marcus Wanselius
- Department of Medicinal Chemistry, Uppsala University, BMC P.O. Box 574, SE-751 23, Uppsala, Sweden
| | - Susanna Abrahmsén-Alami
- Innovation Strategy & External Liaison. Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Belal I Hanafy
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge, United Kingdom
| | - Mariarosa Mazza
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge, United Kingdom
| | - Per Hansson
- Department of Medicinal Chemistry, Uppsala University, BMC P.O. Box 574, SE-751 23, Uppsala, Sweden.
| |
Collapse
|
2
|
Wang H, de Lucio M, Hu T, Leng Y, Gomez H. A MPET 2-mPBPK model for subcutaneous injection of biotherapeutics with different molecular weights: From local scale to whole-body scale. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 260:108543. [PMID: 39671822 DOI: 10.1016/j.cmpb.2024.108543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND AND OBJECTIVE Subcutaneous injection of biotherapeutics has attracted considerable attention in the pharmaceutical industry. However, there is limited understanding of the mechanisms underlying the absorption of drugs with different molecular weights and the delivery of drugs from the injection site to the targeted tissue. METHODS We propose the MPET2-mPBPK model to address this issue. This multiscale model couples the MPET2 model, which describes subcutaneous injection at the local tissue scale from a biomechanical view, with a post-injection absorption model at injection site and a minimal physiologically-based pharmacokinetic (mPBPK) model at whole-body scale. Utilizing the principles of tissue biomechanics and fluid dynamics, the local MPET2 model provides solutions that account for tissue deformation and drug absorption in local blood vessels and initial lymphatic vessels during injection. Additionally, we introduce a model accounting for the molecular weight effect on the absorption by blood vessels, and a nonlinear model accounting for the absorption in lymphatic vessels. The post-injection model predicts drug absorption in local blood vessels and initial lymphatic vessels, which are integrated into the whole-body mPBPK model to describe the pharmacokinetic behaviors of the absorbed drug in the circulatory and lymphatic system. RESULTS We establish a numerical model which links the biomechanical process of subcutaneous injection at local tissue scale and the pharmacokinetic behaviors of injected biotherapeutics at whole-body scale. With the help of the model, we propose an explicit relationship between the reflection coefficient and the molecular weight and predict the bioavalibility of biotherapeutics with varying molecular weights via subcutaneous injection. CONCLUSION The considered drug absorption mechanisms enable us to study the differences in local drug absorption and whole-body drug distribution with varying molecular weights. This model enhances the understanding of drug absorption mechanisms and transport routes in the circulatory system for drugs of different molecular weights, and holds the potential to facilitate the application of computational modeling to drug formulation.
Collapse
Affiliation(s)
- Hao Wang
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA.
| | - Mario de Lucio
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| | - Tianyi Hu
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| | - Yu Leng
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA; Los Alamos National Laboratory, Bikini Atoll Rd, Los Alamos NM 87544, USA
| | - Hector Gomez
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| |
Collapse
|
3
|
Rahimi E, Li C, Zhong X, Shi GH, Ardekani AM. The role of initial lymphatics in the absorption of monoclonal antibodies after subcutaneous injection. Comput Biol Med 2024; 183:109193. [PMID: 39423704 DOI: 10.1016/j.compbiomed.2024.109193] [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: 03/06/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/21/2024]
Abstract
The subcutaneous injection is the most common method of administration of monoclonal antibodies (mAbs) due to the patient's comfort and cost-effectiveness. However, the available knowledge about the transport and absorption of this type of biotherapeutics after subcutaneous injection is limited. Here, a mathematical framework to study the subcutaneous drug delivery of mAbs from injection to lymphatic uptake is presented. A poro-hyperelastic model of the tissue is exploited to find the biomechanical response of the tissue together with a transport model based on an advection-diffusion equation in large-deformation poro-hyperelastic Media. The process of mAbs transport to the lymphatic system has two major parts. First is the initial phase, where mAbs are dispersed in the tissue due to momentum exerted by injection. This stage lasts for only a few minutes after the injection. Then there is the second stage, which can take tens of hours, and as a result, mAb molecules are transported from the subcutaneous layer towards initial lymphatics in the dermis to enter the lymphatic system. In this study, we investigate both stages. The process of plume formation, interstitial pressure, and velocity development is explored. Then, the effect of the injection delivery parameters, injection site, and sensitivity of long-term lymphatic uptake due to variability in permeability, diffusivity, viscosity, and binding of mAbs are investigated. Finally, we study two different injection scenarios with variable injection volume and drug concentration inside the syringe and evaluate them based on the rate of lymphatic uptake. We use our results to find an equivalent lymphatic uptake coefficient similar to the coefficient widely used in pharmacokinetic (PK) models to study the absorption of mAbs. Ultimately, we validate our computational model against available experiments in the literature.
Collapse
Affiliation(s)
- Ehsan Rahimi
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Chenji Li
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Xiaoxu Zhong
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
4
|
Gresham J, Bruin G, Picci M, Bechtold-Peters K, Dimke T, Davies E, Błażejczyk K, Willekens W, Fehervary H, Velde GV. Visualisation and quantification of subcutaneous injections of different volumes, viscosities and injection rates: An ex-vivo micro-CT study. J Pharm Sci 2024; 113:3447-3456. [PMID: 39306036 DOI: 10.1016/j.xphs.2024.08.019] [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: 05/07/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 10/11/2024]
Abstract
The effects of subcutaneous (SC) injection parameters such as drug formulation volume, viscosity and injection rate on therapeutic performance and tolerability have not been established for any drug product. In this study four groups of SC injections were performed on fresh ex vivo minipig abdominal tissue samples, varying volume (0.5-1 mL), viscosity (1-11 cP) and rate (0.02-0.1 mL/s). Micro-CT provided high resolution (50 micron) imaging of the SC tissues before and after injection, enabling a detailed 3D visualisation and analysis of how both injection parameters and tissue microstructure influence spatial distribution of injectables. We found that volume was the only significant factor for spatial distribution of injectate within our design space, and there were no significant factors for tissue backpressure. Variability within test groups was typically greater than differences between group means. Accordingly, whilst the higher viscosity formulations consistently exhibited reduced spatial distribution, the sample size was not large enough to establish confidence in this result. Comparing our findings to clinical evidence, we conclude that injection site and depth are more likely to influence PK and bioavailability than volume, viscosity and rate within our experimental space.
Collapse
Affiliation(s)
| | - Gerard Bruin
- Pharmacokinetic Sciences, Novartis Biomedical Research, Basel, Switzerland
| | - Marie Picci
- Clinical Development Excellence, Global Drug Development, Novartis Pharma AG, Switzerland
| | | | - Thomas Dimke
- Pharmacokinetic Sciences, Novartis Biomedical Research, Basel, Switzerland
| | | | - Kasia Błażejczyk
- Molecular Small Animal Imaging Center (MoSAIC), Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Wouter Willekens
- FIBER, KU Leuven Core Facility for Biomechanical Experimentation, Leuven, Belgium
| | - Heleen Fehervary
- FIBER, KU Leuven Core Facility for Biomechanical Experimentation, Leuven, Belgium; Biomechanics Section, Mechanical Engineering Department, KU Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Molecular Small Animal Imaging Center (MoSAIC), Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium; Biomedical MRI, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Banks D, Kempf JG, Du Y, Reichert P, Narasimhan C, Fang R, Kwon S, Ling J, Lay-Fortenbery A, Zhang Y, Ni QZ, Cote A, Su Y. Investigation of Protein Therapeutics in Frozen Conditions Using DNP MAS NMR: A Study on Pembrolizumab. Mol Pharm 2024. [PMID: 39555969 DOI: 10.1021/acs.molpharmaceut.4c00929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The success of modern biopharmaceutical products depends on enhancing the stability of protein therapeutics. Freezing and thawing, which are common thermal stresses encountered throughout the lifecycle of drug substances, spanning protein production, formulation design, manufacturing, storage, and shipping, can impact this stability. Understanding the physicochemical and molecular behaviors of components in biological drug products at temperatures relevant to manufacturing and shipping is essential for assessing stability risks and determining appropriate storage conditions. This study focuses on the stability of high-concentration monoclonal antibody (mAb) pembrolizumab, the drug substance of Keytruda (Merck & Co., Inc., Rahway, NJ, United States), and its excipients in a frozen solution. By leveraging dynamic nuclear polarization (DNP), we achieve more than 100-fold signal enhancements in solid-state NMR (ssNMR), enabling efficient low-temperature (LT) analysis of pembrolizumab without isotopic enrichment. Through both ex situ and in situ ssNMR experiments conducted across a temperature range of 297 to 77 K, we provide insights into the stability of crystalline pembrolizumab under frozen conditions. Importantly, utilizing LT magic-angle spinning (MAS) probes allows us to study molecular dynamics in pembrolizumab from room temperature down to liquid nitrogen temperatures (<100 K). Our results demonstrate that valuable insights into protein conformation and dynamics, crystallinity, and the phase transformations of excipients during the freezing of the formulation matrix can be readily obtained for biological drug products. This study underscores the potential of LT-MAS ssNMR and DNP techniques for analyzing protein therapeutics and vaccines in frozen solutions.
Collapse
Affiliation(s)
- Daniel Banks
- Bruker Biospin Corporation, Billerica, Massachusetts 01821, United States
| | - James G Kempf
- Bruker Biospin Corporation, Billerica, Massachusetts 01821, United States
| | - Yong Du
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Paul Reichert
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Chakravarthy Narasimhan
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Rui Fang
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Soonbum Kwon
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jing Ling
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ashley Lay-Fortenbery
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yongqian Zhang
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Qing Zhe Ni
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Aaron Cote
- Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yongchao Su
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
6
|
Milewski M, Murashov M, Kapoor Y, Zhang J, Zhu W, Cueto MA, Buist N. Predicting Human Subcutaneous Bioavailability of Therapeutic Monoclonal Antibodies from Systemic Clearance and Volume of Distribution. Mol Pharm 2024; 21:4947-4959. [PMID: 39226331 DOI: 10.1021/acs.molpharmaceut.4c00132] [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] [Indexed: 09/05/2024]
Abstract
Subcutaneous delivery of monoclonal antibody therapeutics is often preferred to intravenous delivery due to better patient compliance and overall lower cost to the healthcare system. However, the systemic absorption of biologics dosed subcutaneously is often incomplete. The aim of this work was to describe a human bioavailability prediction method for monoclonal antibodies delivered subcutaneously that utilizes intravenous pharmacokinetic parameters as input. A two-compartment pharmacokinetic model featuring a parallel-competitive absorption pathway and a presystemic metabolism pathway was employed. A training data set comprised 19 monoclonal antibodies (geometric mean bioavailability of 68%), with previously reported human pharmacokinetic parameters, while a validation set included data compiled from 5 commercial drug products (geometric mean bioavailability of 69%). A single fitted absorption rate constant, paired with compound-specific estimates of presystemic metabolism rate proportional to compound-specific systemic clearance parameters, resulted in calculations of human subcutaneous bioavailability closely mimicking clinical data in the training data set with a root-mean-square error of 5.5%. Application of the same approach to the validation data set resulted in predictions characterized by 12.6% root-mean-square error. Factors that may have impacted the prediction accuracy include a limited number of validation data set compounds and an uncertainty in the absorption rate, which were subsequently discussed. The predictive method described herein provides an initial estimate of the subcutaneous bioavailability based exclusively on pharmacokinetic parameters available from intravenous dosing.
Collapse
Affiliation(s)
- Mikolaj Milewski
- Merck & Co., Inc. 126 East Lincoln Avenue, P.O. Box 2000 Rahway New Jersey 07065 United States
| | - Mikhail Murashov
- Merck & Co., Inc. 126 East Lincoln Avenue, P.O. Box 2000 Rahway New Jersey 07065 United States
| | - Yash Kapoor
- Merck & Co., Inc. 126 East Lincoln Avenue, P.O. Box 2000 Rahway New Jersey 07065 United States
| | - Jingtao Zhang
- Merck & Co., Inc. 126 East Lincoln Avenue, P.O. Box 2000 Rahway New Jersey 07065 United States
| | - Wei Zhu
- Merck & Co., Inc. 126 East Lincoln Avenue, P.O. Box 2000 Rahway New Jersey 07065 United States
| | - Maria A Cueto
- Merck & Co., Inc. 126 East Lincoln Avenue, P.O. Box 2000 Rahway New Jersey 07065 United States
| | - Nicole Buist
- Merck & Co., Inc. 126 East Lincoln Avenue, P.O. Box 2000 Rahway New Jersey 07065 United States
| |
Collapse
|
7
|
Cui C, Wang J, Wang C, Xu T, Qin L, Xiao S, Gong J, Song L, Liu D. Model-informed drug development of envafolimab, a subcutaneously injectable PD-L1 antibody, in patients with advanced solid tumors. Oncologist 2024; 29:e1189-e1200. [PMID: 38982653 PMCID: PMC11379657 DOI: 10.1093/oncolo/oyae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/17/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Envafolimab is the first and only globally approved subcutaneously injectable PD-L1 antibody for the treatment of instability-high (MSI-H) or DNA mismatch repair deficient (dMMR) advanced solid tumors in adults, including those with advanced colorectal cancer that has progressed after treatment with a fluoropyrimidine, oxaliplatin, and irinotecan. The aim of this investigation was to examine the pharmacokinetic and exposure-response (E-R) profile of envafolimab in patients with solid tumors to support the approval of fixed and alternative dose regimens. METHODS In this study, a population pharmacokinetic (PopPK) modeling approach will be employed to quantitatively evaluate intrinsic and extrinsic covariates. Additionally, PopPK-estimated exposure parameters were used to evaluate E-R relationship for safety and efficacy to provide a theoretical basis for recommending optimal treatment regimens. Simulations were performed on the dosing regimens of body weight-based regimen of 2.50 mg/kg QW, fixed dose 150 mg QW, and 300 mg Q2W for the selection of alternative dosing regimens. Data from 4 clinical studies (NCT02827968, NCT03101488, NCT03248843, and NCT03667170) were utilized. RESULTS The PopPK dataset comprised 182 patients with 1810 evaluable envafolimab concentration records. Finally, a one-compartment model incorporating first-order absorption, first-order linear elimination, and time-dependent elimination according to an Emax function was found to accurately describe the concentration-time data of envafolimab in patients with advanced solid tumors. Creatinine clearance and country were identified as statistically significant factors affecting clearance, but had limited clinical significance. A relative flat exposure-response relationship was observed between early measures of safety and efficacy to verify that no dose adjustment is required. Simulation results indicated that 2.50 mg/kg QW, 150 mg QW, and 300 mg Q2W regimen yield similar steady-state exposure. CONCLUSIONS No statistically significant difference was observed between weight-based and fixed dose regimens. Model-based simulation supports the adoption of a 150 mg weekly or 300 mg biweekly dosing regimen of envafolimab in the solid tumor population, as these schedules effectively balance survival benefits and safety risks.
Collapse
Affiliation(s)
- Cheng Cui
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Jing Wang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Chunyang Wang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Ting Xu
- Alphamab Co., Ltd., Suzhou, People’s Republic of China
| | - Lan Qin
- 3DMedicines Co., Ltd., Shanghai, People’s Republic of China
| | - Shen Xiao
- 3DMedicines Co., Ltd., Shanghai, People’s Republic of China
| | - John Gong
- 3DMedicines Co., Ltd., Shanghai, People’s Republic of China
| | - Ling Song
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Somby K, Campagna V, Sánchez-Félix MV, Forbes B, Vllasaliu D. Development of in vitro biopharmaceutics tools for predicting the bioavailability of subcutaneously injected monoclonal antibodies and oligonucleotides. Expert Opin Drug Deliv 2024; 21:1307-1310. [PMID: 39279472 DOI: 10.1080/17425247.2024.2403469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Affiliation(s)
- Karin Somby
- King's College London, Institute of Pharmaceutical Science, London, England
- Novartis Pharma AG, Basel, Switzerland
| | | | - Manuel V Sánchez-Félix
- Formerly with Novartis Pharma AG, Cambridge, USA
- Currently with Halozyme Therapeutics, San Diego, CA, USA
| | - Ben Forbes
- King's College London, Institute of Pharmaceutical Science, London, England
| | - Driton Vllasaliu
- King's College London, Institute of Pharmaceutical Science, London, England
| |
Collapse
|
9
|
Jin SE, Kim J, Sung JH. Recent approaches of antibody therapeutics in androgenetic alopecia. Front Pharmacol 2024; 15:1434961. [PMID: 39221145 PMCID: PMC11362041 DOI: 10.3389/fphar.2024.1434961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Therapeutic antibodies (Abs) have been anticipated as promising alternatives to conventional treatments such as topical minoxidil and oral finasteride for androgenetic alopecia (AGA). Due to the high molecular weight of typical Abs, the half-life of subcutaneous Abs exceeds 2 weeks, allowing an administration intervals of once a month or longer. Direct injection into the areas of hair loss is also feasible, potentially enhancing treatment efficacy while minimizing systemic side effects. However, therapeutic Abs are rarely developed for AGA therapy due to the requirement to be responsiveness to androgens and to exist in the extracellular fluid or cell surface surrounding the hair follicle. In this review, we introduce recent progress of antibody therapeutics in AGA targeting the prolactin receptor, Interleukin-6 receptor, C-X-C motif chemokine ligand 12, and dickkopf 1. As therapeutic Abs for AGA are still in the early stages, targets need further validation and optimization for clinical application.
Collapse
Affiliation(s)
- Su-Eon Jin
- Epi Biotech Co., Ltd., Incheon, Republic of Korea
| | - Jino Kim
- New Hair Plastic Surgery Clinic, Seoul, Republic of Korea
| | | |
Collapse
|
10
|
Stader F, Liu C, Derbalah A, Momiji H, Pan X, Gardner I, Jamei M, Sepp A. A Physiologically Based Pharmacokinetic Model Relates the Subcutaneous Bioavailability of Monoclonal Antibodies to the Saturation of FcRn-Mediated Recycling in Injection-Site-Draining Lymph Nodes. Antibodies (Basel) 2024; 13:70. [PMID: 39189241 PMCID: PMC11348173 DOI: 10.3390/antib13030070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
The bioavailability of a monoclonal antibody (mAb) or another therapeutic protein after subcutaneous (SC) dosing is challenging to predict from first principles, even if the impact of injection site physiology and drug properties on mAb bioavailability is generally understood. We used a physiologically based pharmacokinetic model to predict pre-systemic clearance after SC administration mechanistically by incorporating the FcRn salvage pathway in antigen-presenting cells (APCs) in peripheral lymph nodes, draining the injection site. Clinically observed data of the removal rate of IgG from the arm as well as its plasma concentration after SC dosing were mostly predicted within the 95% confidence interval. The bioavailability of IgG was predicted to be 70%, which mechanistically relates to macropinocytosis in the draining lymph nodes and transient local dose-dependent partial saturation of the FcRn receptor in the APCs, resulting in higher catabolism and consequently less drug reaching the systemic circulation. The predicted free FcRn concentration was reduced to 40-45%, reaching the minimum 1-2 days after the SC administration of IgG, and returned to baseline after 8-12 days, depending on the site of injection. The model predicted the uptake into APCs, the binding affinity to FcRn, and the dose to be important factors impacting the bioavailability of a mAb.
Collapse
Affiliation(s)
- Felix Stader
- Simcyp Division, Certara UK Ltd., Level 2 Acero, 1 Concourse Way, Sheffield S1 2BJ, UK (X.P.); (I.G.); (A.S.)
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Ait‐Oudhia S, Jaworowicz D, Hu Z, Bihorel S, Hu S, Balasubrahmanyam B, Mistry B, de Oliveira Pena J, Wenning L, Gheyas F. Population pharmacokinetic modeling of sotatercept in healthy participants and patients with pulmonary arterial hypertension. CPT Pharmacometrics Syst Pharmacol 2024; 13:1380-1393. [PMID: 38812074 PMCID: PMC11330185 DOI: 10.1002/psp4.13166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Sotatercept is a breakthrough, first-in-class biologic, that is FDA-approved for the treatment of pulmonary arterial hypertension (PAH). A population pharmacokinetic (PopPK) model was developed using data from two phase 1 studies in healthy participants, and two phase 2 studies and one phase 3 study in participants with PAH. The pooled sotatercept PK data encompassed single intravenous (IV) or subcutaneous (SC) doses ranging from 0.01 to 3.0 mg/kg, as well as multiple SC doses ranging from 0.03 to 1.0 mg/kg, with PK samples collected up to a maximum of ~150 weeks following Q3W and Q4W dosing regimens. The final PopPK analysis included 350 participants, with 30 and 320 participants receiving sotatercept IV and SC, respectively. A two-compartment model with a first-order absorption rate constant and a linear disposition from central compartment well-described sotatercept PK. The estimated bioavailability is ~66%; bioavailability, clearance (CL), and central volume (VC) have low to moderate inter-individual variability. Time-varying body weight and baseline albumin concentration were statistically significant predictors of PK; CL and VC were predicted to increase with increasing body weight, while CL was predicted to decrease with increasing baseline albumin concentration. However, the magnitude of covariate effects is not predicted to meaningfully alter the disposition of sotatercept. Altogether, the PopPK modeling results demonstrate favorable PK characteristics (low to moderate variability and typical bioavailability), supporting sotatercept as a SC biological agent for the treatment of patients with PAH.
Collapse
Affiliation(s)
| | | | - Ziheng Hu
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | - Shuai Hu
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | - Bipin Mistry
- Acceleron Pharma, a subsidiary of Merck & Co., Inc.RahwayNew JerseyUSA
| | | | | | | |
Collapse
|
12
|
Novikov N, Buch A, Yang H, Andruk M, Liu G, Wu M, Howell H, MacDonald B, Savage W. First-in-Human Phase 1 Study Evaluating the Safety, Pharmacokinetics, and Pharmacodynamics of DISC-0974, an Anti-Hemojuvelin Antibody, in Healthy Participants. J Clin Pharmacol 2024; 64:953-962. [PMID: 38515275 DOI: 10.1002/jcph.2432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Pathologic elevations in hepcidin, a key regulator of iron homeostasis, contribute to anemia of inflammation in chronic disease. DISC-0974 is a monoclonal antibody that binds to hemojuvelin and blocks bone morphogenetic protein signaling, thereby suppressing hepcidin production. Reduction of systemic hepcidin levels is predicted to increase iron absorption and mobilize stored iron into circulation, where it may be utilized by red blood cell (RBC) precursors in the bone marrow to improve hemoglobin levels and to potentially alleviate anemia of inflammation. We conducted a first-in-human, double-blind, placebo-controlled, single-ascending dose study to evaluate safety, pharmacokinetics, and pharmacodynamics of DISC-0974 in healthy participants. Overall, 42 participants were enrolled and received a single dose of placebo or DISC-0974 at escalating dose levels (7-56 mg), administered intravenously (IV) or subcutaneously (SC). DISC-0974 was well tolerated, with a safety profile comparable to that of placebo. Pharmacokinetic data was dose and route related, with a terminal half-life of approximately 7 days. The bioavailability of SC dosing was ∼50%. Pharmacodynamic data showed dose-dependent decreases in serum hepcidin, with reductions of nearly 75% relative to baseline at the highest dose level tested, and corresponding increases in serum iron in response to DISC-0974 administration. Dose-dependent changes in serum ferritin and hematology parameters were also observed, indicating mobilization of iron stores and downstream effects of enhanced hemoglobinization and production of RBCs. Altogether, these data are consistent with the mechanism of action of DISC-0974 and support the selection of a biologically active dose range for evaluation in clinical trials for individuals with anemia of inflammation.
Collapse
MESH Headings
- Humans
- Male
- Adult
- Double-Blind Method
- Female
- Hepcidins/blood
- Middle Aged
- Healthy Volunteers
- Young Adult
- Hemochromatosis Protein
- GPI-Linked Proteins/antagonists & inhibitors
- Dose-Response Relationship, Drug
- Half-Life
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/pharmacology
- Iron
- Injections, Subcutaneous
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacology
- Adolescent
Collapse
Affiliation(s)
| | - Akshay Buch
- Disc Medicine, Watertown, Massachusetts, USA
| | - Hua Yang
- Disc Medicine, Watertown, Massachusetts, USA
| | | | - Guowen Liu
- Disc Medicine, Watertown, Massachusetts, USA
| | - Min Wu
- Disc Medicine, Watertown, Massachusetts, USA
| | | | | | - Will Savage
- Disc Medicine, Watertown, Massachusetts, USA
| |
Collapse
|
13
|
Martin SJ, Guenette M, Oh J. Evaluating the Therapeutic Potential of Ublituximab in the Treatment of MS: Design, Development and Place in Therapy. Drug Des Devel Ther 2024; 18:3025-3042. [PMID: 39050801 PMCID: PMC11268567 DOI: 10.2147/dddt.s388410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
B cells are critical to the pathogenesis of multiple sclerosis (MS), an autoimmune disease of the central nervous system. B cell depletion using anti-CD20 monoclonal antibodies (mAbs) has proven to be an extremely successful treatment strategy, with profound suppression of both clinical and radiological evidence of focal inflammatory disease. Several anti-CD20 mAbs are now licensed for use in MS, with ublituximab being the latest to gain regulatory approval. The unique properties of each of the anti-CD20 mAb may result in nuanced differences in timing, duration and depth of B cell depletion, with the potential for such differences to have a clinical relevance to both drug efficacy and adverse effects. In this review, we summarize the design, development, and current place in MS therapy for ublituximab.
Collapse
Affiliation(s)
- Sarah-Jane Martin
- Division of Neurology, Department of Medicine, St Michael’s Hospital, Toronto, Canada
- University of Glasgow, Glasgow, UK
| | - Melanie Guenette
- Division of Neurology, Department of Medicine, St Michael’s Hospital, Toronto, Canada
| | - Jiwon Oh
- Division of Neurology, Department of Medicine, St Michael’s Hospital, Toronto, Canada
| |
Collapse
|
14
|
Khan T, Hussain A, Siddique MUM, Altamimi MA, Malik A, Bhat ZR. HSPiP, Computational, and Thermodynamic Model-Based Optimized Solvents for Subcutaneous Delivery of Tolterodine Tartrate and GastroPlus‑Based In Vivo Prediction in Humans: Part II. AAPS PharmSciTech 2024; 25:160. [PMID: 38992299 DOI: 10.1208/s12249-024-02880-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
In part I, we reported Hansen solubility parameters (HSP, HSPiP program), experimental solubility at varied temperatures for TOTA delivery. Here, we studied dose volume selection, stability, pH, osmolality, dispersion, clarity, and viscosity of the explored combinations (I-VI). Ex vivo permeation and deposition studies were performed to observe relative diffusion rate from the injected site in rat skin. Confocal laser scanning microscopy (CLSM) study was conducted to support ex vivo findings. Moreover, GastroPlus predicted in vivo parameters in humans and the impact of various critical factors on pharmacokinetic parameters (PK). Immediate release product (IR) contained 60% of PEG400 whereas controlled release formulation (CR) contained PEG400 (60%), water (10%) and d-limonene (30%) to deliver 2 mg of TOTA. GastroPlus predicted the plasma drug concentration of weakly basic TOTA as function of pH (from pH 2.0 to 9). The cumulative drug permeation and drug deposition were found to be in the order as B-VI˃ C-VI˃A-VI across rat skin. This finding was further supported with CLSM. Moreover, IR and CR were predicted to achieve Cmax of 0.0038 µg/ mL and 0.00023 µg/mL, respectively, after sub-Q delivery. Added limonene in CR extended the plasma drug concentration over period of 12 h as predicted in GastroPlus. Parameters sensitivity analysis (PSA) assessment predicted that sub-Q blood flow rate is the only factor affecting PK parameters in IR formulation whereas this was insignificant for CR. Thus, sub-Q delivery CR would be promising alternative with ease of delivery to children and aged patient.
Collapse
Affiliation(s)
- Tasneem Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Afzal Hussain
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Mohd Usman Mohd Siddique
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy Dhule, Dhule, MH, 424001, India
| | - Mohammad A Altamimi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Zahid Rafiq Bhat
- Department of Molecular and Cellular Oncology, MD Anderson Cancer Centre, Houston, Texas, USA
| |
Collapse
|
15
|
Kumar M, Lanke S, Yadav A, Ette M, Mager DE, Shah DK. Inter-Antibody Variability in the Clinical Pharmacokinetics of Monoclonal Antibodies Characterized Using Population Physiologically Based Pharmacokinetic Modeling. Antibodies (Basel) 2024; 13:54. [PMID: 39051330 PMCID: PMC11270311 DOI: 10.3390/antib13030054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/27/2024] Open
Abstract
The objective of this work was to develop a population physiologically based pharmacokinetic (popPBPK) model to characterize the variability in the clinical PK of monoclonal antibodies (mAbs) following intravenous (IV) and subcutaneous (SC) administration. An extensive literature search was conducted and clinical PK data for FDA-approved as well as non-approved mAbs were collected. Training and validation datasets of 44 and 9 mAbs exhibiting linear pharmacokinetics were used for model development. The variability in antibody PK was captured by accounting for different rate constants of pinocytosis (CLup) and intracellular degradation (kdeg) for different mAbs. Typical values for CLup and kdeg and their respective inter-antibody variabilities (ωClup, ωKdeg) were estimated to be 0.32 L/h/L and 26.1 h-1 (73% and 46%). Varied absorption profiles following SC dosing were characterized by incorporating inter-antibody variability in local degradation (kSC) and rate of lymphatic uptake (S_Lu) of mAbs. Estimates for typical kSC and S_Lu values, and ωKsc,ωS_Lu, were found to be 0.0015 h-1 and 0.54 (193%, and 49%). FDA-approved mAbs showed less local degradation (0.0014 h-1 vs. 0.0038 h-1) compared with other clinically tested mAbs, whereas no substantial differences in physiological processes involved in disposition were observed. To evaluate the generalizability of estimated PK parameters and model validation, the final popPBPK model was used to simulate the range of expected PK for mAbs following SC administration of nine different mAbs that were not used for model-building purposes. The predicted PK of all nine mAbs was within the expected range specified a priori. Thus, the popPBPK model presented here may serve as a tool to predict the clinical PK of mAbs with linear disposition before administering them to humans. The model may also support preclinical-to-clinical translation and 'first-in-human' dose determination for mAbs.
Collapse
Affiliation(s)
- Mokshada Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Sravani Lanke
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Alka Yadav
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Mfonabasi Ette
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Donald E. Mager
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
- Enhanced Pharmacodynamics, LLC, Buffalo, NY 14203, USA
| | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| |
Collapse
|
16
|
Scavone C, Anatriello A, Baccari I, Cantone A, Di Giulio Cesare D, Bernardi FF, Moreggia O, Liguori V, Andreone V, Maniscalco GT, Capuano A. Comparison of injective related reactions following ofatumumab and ocrelizumab in patients with multiple sclerosis: data from the European spontaneous reporting system. Front Neurol 2024; 15:1383910. [PMID: 38994488 PMCID: PMC11236557 DOI: 10.3389/fneur.2024.1383910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction In 2021 ofatumumab, a recombinant human anti-CD20 monoclonal antibody (mAb) already authorized for the treatment of chronic lymphocytic leukemia, received the marketing approval for the treatment of relapsing forms of multiple sclerosis (MS). Differently from ocrelizumab, that is administered intravenously, ofatumumab if the first anti-CD20 mAb to be administered subcutaneously without a premedication. Methods and objectives In this study we aimed to describe and compare the main characteristics of Individual Case Safety Reports (ICSRs) describing the occurrence of Injective Related Reactions (IRRs) following the treatment with ocrelizumab and ofatumumab reported in the Eudravigilance (EV) database during years 2021-2023. Results A total of 860 ICSRs with either ofatumumab and ocrelizumab as suspected drug were retrieved from Eudravigilance, of which 51% associated with ofatumumab and 49% with ocrelizumab. The majority of patients who experienced IRRs following ocrelizumab belonged to the age group of 18-64 years (73%), while the age-group was mostly not specified (55%) in ICSRs reporting ofatumumab as suspected. The distribution of gender was almost similar in the two groups, with the majority of ICSRs related to female patients. "Pyrexia" was the Preferred Term (PT) most reported for ofatumumab, while "Infusion related reaction" were more frequently reported with ocrelizumab. Premedication drugs were reported in 148 ICSRs. Out of 89 ICSRs for which the Time to Event (TTE) was calculated, 74 reported IRRs that occurred the same day of the drug administration. Discussion Based on the results of this study, although a risk of ofatumumab-induced IRRs cannot be excluded, it should be considered as manageable considering that the drug seems to be mostly associated with the occurrence of fever. Thus, it is important to continue to closely monitor the use of these in clinical practice to improve the knowledge on their long-term safety.
Collapse
Affiliation(s)
- Cristina Scavone
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Regional Center of Pharmacovigilance and Pharmacoepidemiology of Campania Region, Naples, Italy
| | - Antonietta Anatriello
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Regional Center of Pharmacovigilance and Pharmacoepidemiology of Campania Region, Naples, Italy
| | - Isabella Baccari
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Regional Center of Pharmacovigilance and Pharmacoepidemiology of Campania Region, Naples, Italy
| | - Andrea Cantone
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Regional Center of Pharmacovigilance and Pharmacoepidemiology of Campania Region, Naples, Italy
| | | | | | - Ornella Moreggia
- Multiple Sclerosis Regional Center, "A. Cardarelli" Hospital, Naples, Italy
| | - Valerio Liguori
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Regional Center of Pharmacovigilance and Pharmacoepidemiology of Campania Region, Naples, Italy
| | - Vincenzo Andreone
- Neurological Clinic and Stroke Unit, "A. Cardarelli" Hospital, Naples, Italy
| | - Giorgia Teresa Maniscalco
- Multiple Sclerosis Regional Center, "A. Cardarelli" Hospital, Naples, Italy
- Neurological Clinic and Stroke Unit, "A. Cardarelli" Hospital, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Regional Center of Pharmacovigilance and Pharmacoepidemiology of Campania Region, Naples, Italy
| |
Collapse
|
17
|
Torres-Terán I, Venczel M, Klein S. Prediction of subcutaneous drug absorption - Development of novel simulated interstitial fluid media for predictive subcutaneous in vitro assays. Int J Pharm 2024; 658:124227. [PMID: 38750979 DOI: 10.1016/j.ijpharm.2024.124227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
Media that mimic physiological fluids at the site of administration have proven to be valuable in vitro tools for predicting in vivo drug release, particularly for routes of administration where animal studies cannot accurately predict human performance. The objective of the present study was to develop simulated interstitial fluids (SISFs) that mimic the major components and physicochemical properties of subcutaneous interstitial fluids (ISFs) from preclinical species and humans, but that can be easily prepared in the laboratory and used in in vitro experiments to estimate in vivo drug release and absorption of subcutaneously administered formulations. Based on data from a previous characterization study of ISFs from different species, two media were developed: a simulated mouse-rat ISF and a simulated human-monkey ISF. The novel SISFs were used in initial in vitro diffusion studies with a commercial injectable preparation of liraglutide. Although the in vitro model used for this purpose still requires significant refinement, these two new media will undoubtedly contribute to a better understanding of the in vivo performance of subcutaneous injectables in different species and will help to reduce the number of unnecessary in vivo experiments in preclinical species by implementation in predictive in vitro models.
Collapse
Affiliation(s)
- Iria Torres-Terán
- University of Greifswald. Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, 3 Felix Hausdorff Street, 17489 Greifswald, Germany; Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt am Main, Germany
| | - Márta Venczel
- University of Greifswald. Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, 3 Felix Hausdorff Street, 17489 Greifswald, Germany
| | - Sandra Klein
- Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt am Main, Germany.
| |
Collapse
|
18
|
Gomes C, Gridley K, Anastasiou I, Sinkó B, Mrsny RJ. Hydrogel formats to model potential drug interactions occurring at the subcutaneous injection site. Eur J Pharm Biopharm 2024; 199:114308. [PMID: 38688439 DOI: 10.1016/j.ejpb.2024.114308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/13/2024] [Accepted: 04/28/2024] [Indexed: 05/02/2024]
Abstract
We have previously developed an in vitro instrument, termed subcutaneous injection site simulator (SCISSOR), that can be used to monitor release properties of an active pharmaceutical ingredient (API) and formulation components of a medicine designed for SC injection. Initial studies to validate the SCISSOR instrument applications used a simple hyaluronic acid (HA) hydrogel to monitor early release events. We now report a type of cross-linked HA that can, when combined with HA, provide a hydrogel (HA-XR) with optical clarity and rheological properties that remain stable for at least 6 days. Incorporation of 0.05-0.1 mg/mL of collagens isolated from human fibroblasts (Col F), bovine type I collagen (Col I), chicken collagen type II (Col II), or chondroitin sulphate (CS) produced HA or HA-XR hydrogel formats with optical clarity and rheological properties comparable to HA or HA-XR alone. HA + Col F hydrogel had a much greater effect on release rates of 70 kDa compared to 4 kDa dextran, while Col F incorporated into the HA-XR hydrogel accentuated differences in release rates of prandial and basal forms of insulin as well as decreased the release rate of denosumab. A hydrogel format of HA + Col I was used to examine the complex events for bevacizumab release under conditions where a target ligand (vascular endothelial growth factor) can interact with extracellular matrix (ECM). Together, these data have demonstrated the feasibility of using a cross-linked HA format to examine API release over multiple days and incorporation of specific ECM elements to prepare more biomimetic hydrogels that allow for tractable examination of their potential impact of API release.
Collapse
Affiliation(s)
| | - Kate Gridley
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | | | | | - Randall J Mrsny
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK.
| |
Collapse
|
19
|
Mathias N, Huille S, Picci M, Mahoney RP, Pettis RJ, Case B, Helk B, Kang D, Shah R, Ma J, Bhattacharya D, Krishnamachari Y, Doucet D, Maksimovikj N, Babaee S, Garidel P, Esfandiary R, Gandhi R. Towards more tolerable subcutaneous administration: Review of contributing factors for improving combination product design. Adv Drug Deliv Rev 2024; 209:115301. [PMID: 38570141 DOI: 10.1016/j.addr.2024.115301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Subcutaneous (SC) injections can be associated with local pain and discomfort that is subjective and may affect treatment adherence and overall patient experience. With innovations increasingly focused on finding ways to deliver higher doses and volumes (≥2 mL), there is a need to better understand the multiple intertwined factors that influence pain upon SC injection. As a priority for the SC Drug Development & Delivery Consortium, this manuscript provides a comprehensive review of known attributes from published literature that contribute to pain/discomfort upon SC injection from three perspectives: (1) device and delivery factors that cause physical pain, (2) formulation factors that trigger pain responses, and (3) human factors impacting pain perception. Leveraging the Consortium's collective expertise, we provide an assessment of the comparative and interdependent factors likely to impact SC injection pain. In addition, we offer expert insights and future perspectives to fill identified gaps in knowledge to help advance the development of patient-centric and well tolerated high-dose/high-volume SC drug delivery solutions.
Collapse
Affiliation(s)
- Neil Mathias
- Bristol-Myers Squibb, Co., 1 Squibb Dr, New Brunswick, NJ, 08901 USA
| | - Sylvain Huille
- Sanofi, 13 quai Jules Guesde, 94400 Vitry-Sur-Seine, France.
| | - Marie Picci
- Novartis Pharma AG, Fabrikstrasse 4, CH-4056 Basel, Switzerland
| | - Robert P Mahoney
- Comera Life Sciences, 12 Gill St, Suite 4650, Woburn, MA 01801 USA
| | - Ronald J Pettis
- Becton-Dickinson, 21 Davis Drive, Research Triangle Park, NC 27513 USA
| | - Brian Case
- KORU Medical Systems, 100 Corporate Dr, Mahwah, NJ 07430 USA
| | - Bernhard Helk
- Novartis Pharma AG, Werk Klybeck, WKL-681.4.42, CH-4057 Basel, Switzerland
| | - David Kang
- Halozyme Therapeutics, Inc., 12390 El Camino Real, San Diego, CA 92130 USA
| | - Ronak Shah
- Bristol-Myers Squibb, Co., 1 Squibb Dr, New Brunswick, NJ, 08901 USA
| | - Junchi Ma
- Johnson & Johnson Innovative Medicine, 200 Great Valley Pkwy, Malvern, PA 19355 USA
| | | | | | - Dany Doucet
- GSK, 1250 South Collegeville Road, Collegeville, PA 19426 USA
| | | | - Sahab Babaee
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065 USA
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397 Biberach/Riss, Germany
| | | | - Rajesh Gandhi
- Bristol-Myers Squibb, Co., 1 Squibb Dr, New Brunswick, NJ, 08901 USA
| |
Collapse
|
20
|
Lin KJ, Turner KC, Hassan HE, Harnisch LO, Davis JD, DiCioccio AT. Population Pharmacokinetics of Fasinumab in Healthy Volunteers and Patients With Pain Due to Osteoarthritis of the Knee or Hip. Clin Pharmacol Drug Dev 2024; 13:621-630. [PMID: 38323727 DOI: 10.1002/cpdd.1380] [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: 10/26/2023] [Accepted: 12/27/2023] [Indexed: 02/08/2024]
Abstract
Osteoarthritis (OA) pain management options are currently limited. Fasinumab, an anti-nerve growth factor monoclonal antibody, has been investigated in healthy volunteers and patients with OA-related pain, among other conditions. Data from 12 Phase I-III clinical trials of 92 healthy volunteers and 7430 patients with OA were used to develop a population pharmacokinetic model to characterize fasinumab concentration-time profiles and assess the covariates' effect on fasinumab pharmacokinetic parameters. Participants received single or repeated fasinumab doses intravenously (IV)/subcutaneously (SC), based on body weight (0.03-1 mg/kg IV or 0.1-0.3 mg/kg SC)/fixed dose (9-12 mg IV or 1-12 mg SC). Fasinumab concentration-time data following IV and SC administration in healthy volunteers and patients with OA-related pain were adequately described by a 2-compartment model. Bioavailability increased with higher doses; estimated at 55.1% with 1 mg SC dose, increasing in a greater-than-proportional manner above this. Body weight had the largest predicted impact on fasinumab steady-state exposures, participants at the 5th and 95th percentiles had a 43%-45% higher/22%-23% lower exposure versus reference, respectively. Other covariates had small but clinically irrelevant impacts.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Adult
- Osteoarthritis, Knee/drug therapy
- Osteoarthritis, Knee/complications
- Healthy Volunteers
- Aged
- Osteoarthritis, Hip/drug therapy
- Osteoarthritis, Hip/complications
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Models, Biological
- Pain/drug therapy
- Biological Availability
- Injections, Subcutaneous
- Young Adult
- Dose-Response Relationship, Drug
- Clinical Trials, Phase III as Topic
Collapse
Affiliation(s)
- Kuan-Ju Lin
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | - John D Davis
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | |
Collapse
|
21
|
Stevenson J, Poker R, Schoss J, Campbell M, Everitt C, Holly B, Stones N, Pettis RJ, Sanchez-Felix M. Pharmaceutical and biotech industry perspectives on optimizing patient experience and treatment adherence through subcutaneous drug delivery design. Adv Drug Deliv Rev 2024; 209:115322. [PMID: 38677443 DOI: 10.1016/j.addr.2024.115322] [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: 02/02/2024] [Revised: 04/06/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
Subcutaneous (SC) drug delivery can be a safe, effective alternative to the traditional intravenous route of administration, potentially offering notable advantages for both patients and healthcare providers. The SC Drug Development & Delivery Consortium convened in 2018 to raise awareness of industry challenges to advance the development of patient-centric SC drug delivery strategies. The SC Consortium identified better understanding of patient preferences and perspectives as necessary to optimize SC product design attributes and help guide design decisions during SC product development. This manuscript provides a comprehensive overview of patient-centric factors for consideration in the SC drug delivery design and development process with the aim of establishing a foundation of existing knowledge for patient experiences related to SC drug delivery. This overview is informed by the outcomes of a multi-step survey of Consortium members and key pharmaceutical stakeholders. Framed in the context of the patient's treatment journey, the survey findings offer future perspectives to fill data gaps to advance patient-centric SC drug delivery.
Collapse
Affiliation(s)
| | - Rachel Poker
- AstraZeneca, Human Factors Engineering, BioPharmaceutical Development, Biopharmaceuticals R&D, 121 Oyster Point Blvd, South San Francisco, CA 94080, USA
| | | | | | - Claire Everitt
- Pfizer, Granta Park, Great Abington, Cambridge CB21 6GP, UK
| | - Brian Holly
- Pfizer, Granta Park, Great Abington, Cambridge CB21 6GP, UK
| | - Nicholas Stones
- Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland
| | - Ronald J Pettis
- Becton-Dickinson, 21 Davis Drive, Research Triangle Park, NC 27513, USA
| | | |
Collapse
|
22
|
Ling SF, Ogungbenro K, Darwich AS, Ariff ABM, Nair N, Bluett J, Morgan AW, Isaacs JD, Wilson AG, Hyrich KL, Barton A, Plant D. Population Pharmacokinetic Analysis and Simulation of Alternative Dosing Regimens for Biosimilars to Adalimumab and Etanercept in Patients with Rheumatoid Arthritis. Pharmaceutics 2024; 16:702. [PMID: 38931826 PMCID: PMC11206727 DOI: 10.3390/pharmaceutics16060702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Efficacy to biologics in rheumatoid arthritis (RA) patients is variable and is likely influenced by each patient's circulating drug levels. Using modelling and simulation, the aim of this study was to investigate whether adalimumab and etanercept biosimilar dosing intervals can be altered to achieve therapeutic drug levels at a faster/similar time compared to the recommended interval. RA patients starting subcutaneous Amgevita or Benepali (adalimumab and etanercept biosimilars, respectively) were recruited and underwent sparse serum sampling for drug concentrations. Drug levels were measured using commercially available kits. Pharmacokinetic data were analysed using a population approach (popPK) and potential covariates were investigated in models. Models were compared using goodness-of-fit criteria. Final models were selected and used to simulate alternative dosing intervals. Ten RA patients starting the adalimumab biosimilar and six patients starting the etanercept biosimilar were recruited. One-compartment PK models were used to describe the popPK models for both drugs; no significant covariates were found. Typical individual parameter estimates were used to simulate altered dosing intervals for both drugs. A simulation of dosing the etanercept biosimilar at a lower rate of every 10 days reached steady-state concentrations earlier than the usual dosing rate of every 7 days. Simulations of altered dosing intervals could form the basis for future personalised dosing studies, potentially saving costs whilst increasing efficacy.
Collapse
Affiliation(s)
- Stephanie F. Ling
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester M13 9PT, UK
| | - Adam S. Darwich
- Division of Health Informatics and Logistics, KTH Royal Institute of Technology, 11428 Stockholm, Sweden
| | - Amirah Binti Mohammad Ariff
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester M13 9PT, UK
| | - Nisha Nair
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - James Bluett
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Ann W. Morgan
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds LS7 4SA, UK
- NIHR In Vitro Diagnostic Co-Operative, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| | - John D. Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK
- Musculoskeletal Unit, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne NE7 7DN, UK
| | - Anthony G. Wilson
- School of Medicine and Medical Science, Conway Institute, University College Dublin, Dublin 4 Belfield, Ireland
| | - Kimme L. Hyrich
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester M13 9PT, UK
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Darren Plant
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| |
Collapse
|
23
|
Desai M, Kenney J, Pezalla E. Evaluating unmet needs in large-volume subcutaneous drug delivery: U.S. payer perspectives on a novel, large-volume on-body delivery system. Curr Med Res Opin 2024:1-12. [PMID: 38700234 DOI: 10.1080/03007995.2024.2351165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
OBJECTIVE Existing healthcare systems face finite resource allocation and budgetary constraints, resulting in a substantial need for innovative solutions to enhance service delivery at reduced costs. A novel, user-friendly on-body delivery system (OBDS) was developed which enables administration of large-volume subcutaneous (SC) drugs in both clinical and home-based settings (at-home healthcare professional [HCP] administration or at-home self-administration). METHODS This research sought to evaluate the potential economic impact of at-home self- or HCP- administration with the OBDS through a comprehensive review of published literature and semi-structured interviews with 17 US payers representing approximately 227 million covered lives. RESULTS Published literature on OBDS remains limited, but available research highlights the cost-savings of SC administration due to reduced healthcare resource utilization, particularly with home-based care, and improved patient compliance. In interviews, payers identified several attributes that would help address unmet clinical and economic needs. Clinically, the hidden needle and ease-of-use compared to SC syringe pumps was deemed valuable to improve patient compliance and, as OBDS required minimal training, reduce the risk of administration errors. The flexibility to administer drugs at home (self-administration or HCP-administration) or in-clinic was identified as the most impactful attribute on coverage decision making as it has the greatest potential to reduce costs associated with HCP administration for several therapeutic areas. CONCLUSIONS Given the ability to help address critical unmet needs for the patient and healthcare system, a large proportion of the payers stated that the novel OBDS would warrant a price premium versus the cost of the standalone SC vial and certainly over the IV counterpart. Future research to quantify the value that OBDS efficiencies could bring to healthcare delivery are warranted.
Collapse
Affiliation(s)
- Mehul Desai
- Medical Affairs, Enable Injections Inc, Cincinnati, Ohio, USA
| | | | - Edmund Pezalla
- Enlightenment Bioconsult, Wethersfield, Connecticut, USA
| |
Collapse
|
24
|
Du Y, Song J, Lu L, Yeung E, Givand J, Procopio A, Su Y, Hu G. Design of a Reciprocal Injection Device for Stability Studies of Parenteral Biological Drug Products. J Pharm Sci 2024; 113:1330-1338. [PMID: 38113997 DOI: 10.1016/j.xphs.2023.12.014] [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: 10/01/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023]
Abstract
Formulation screening, essential for assessing the impact of physical, chemical, and mechanical stresses on protein stability, plays a critical role in biologics drug product development. This research introduces a Reciprocal Injection Device (RID) designed to accelerate formulation screening by probing protein stability under intensified stress conditions within prefilled syringes. This versatile device is designed to accommodate a broad spectrum of injection parameters and diverse syringe dimensions. A commercial drug product was employed as a model monoclonal antibody formulation. Our findings effectively highlight the efficacy of the RID in assessing concentration-dependent protein stability. This device exhibits significant potential to amplify the influences of interfacial interactions, such as those with buffer salts, excipients, air, metals, and silicone oils, commonly found in combination drug products, and to evaluate the protein stability under varied stresses.
Collapse
Affiliation(s)
- Yong Du
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Jing Song
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Lynn Lu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Edward Yeung
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Jeffrey Givand
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Adam Procopio
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Yongchao Su
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States.
| | - Guangli Hu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States.
| |
Collapse
|
25
|
Mayer BT, Zhang L, deCamp AC, Yu C, Sato A, Angier H, Seaton KE, Yates N, Ledgerwood JE, Mayer K, Caskey M, Nussenzweig M, Stephenson K, Julg B, Barouch DH, Sobieszczyk ME, Edupuganti S, Kelley CF, McElrath MJ, Gelderblom HC, Pensiero M, McDermott A, Gama L, Koup RA, Gilbert PB, Cohen MS, Corey L, Hyrien O, Tomaras GD, Huang Y. Impact of LS Mutation on Pharmacokinetics of Preventive HIV Broadly Neutralizing Monoclonal Antibodies: A Cross-Protocol Analysis of 16 Clinical Trials in People without HIV. Pharmaceutics 2024; 16:594. [PMID: 38794258 PMCID: PMC11125931 DOI: 10.3390/pharmaceutics16050594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/26/2024] Open
Abstract
Monoclonal antibodies are commonly engineered with an introduction of Met428Leu and Asn434Ser, known as the LS mutation, in the fragment crystallizable region to improve pharmacokinetic profiles. The LS mutation delays antibody clearance by enhancing binding affinity to the neonatal fragment crystallizable receptor found on endothelial cells. To characterize the LS mutation for monoclonal antibodies targeting HIV, we compared pharmacokinetic parameters between parental versus LS variants for five pairs of anti-HIV immunoglobin G1 monoclonal antibodies (VRC01/LS/VRC07-523LS, 3BNC117/LS, PGDM1400/LS PGT121/LS, 10-1074/LS), analyzing data from 16 clinical trials of 583 participants without HIV. We described serum concentrations of these monoclonal antibodies following intravenous or subcutaneous administration by an open two-compartment disposition, with first-order elimination from the central compartment using non-linear mixed effects pharmacokinetic models. We compared estimated pharmacokinetic parameters using the targeted maximum likelihood estimation method, accounting for participant differences. We observed lower clearance rate, central volume, and peripheral volume of distribution for all LS variants compared to parental monoclonal antibodies. LS monoclonal antibodies showed several improvements in pharmacokinetic parameters, including increases in the elimination half-life by 2.7- to 4.1-fold, the dose-normalized area-under-the-curve by 4.1- to 9.5-fold, and the predicted concentration at 4 weeks post-administration by 3.4- to 7.6-fold. Results suggest a favorable pharmacokinetic profile of LS variants regardless of HIV epitope specificity. Insights support lower dosages and/or less frequent dosing of LS variants to achieve similar levels of antibody exposure in future clinical applications.
Collapse
Affiliation(s)
- Bryan T. Mayer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Lily Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Allan C. deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Alicia Sato
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Heather Angier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Kelly E. Seaton
- Duke University Medical Center, Durham, NC 27705, USA; (K.E.S.); (N.Y.); (G.D.T.)
| | - Nicole Yates
- Duke University Medical Center, Durham, NC 27705, USA; (K.E.S.); (N.Y.); (G.D.T.)
| | - Julie E. Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | | | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; (M.C.); (M.N.)
| | - Michel Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; (M.C.); (M.N.)
| | - Kathryn Stephenson
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA 02139, USA; (K.S.); (B.J.)
| | - Boris Julg
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA 02139, USA; (K.S.); (B.J.)
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | | | - Srilatha Edupuganti
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.E.); (C.F.K.)
| | - Colleen F. Kelley
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.E.); (C.F.K.)
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Huub C. Gelderblom
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Michael Pensiero
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Adrian McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Lucio Gama
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Myron S. Cohen
- Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
- Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Georgia D. Tomaras
- Duke University Medical Center, Durham, NC 27705, USA; (K.E.S.); (N.Y.); (G.D.T.)
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
26
|
Nolan RP, Printz MA. Modeling the subcutaneous pharmacokinetics of antibodies co-administered with rHuPH20. Clin Transl Sci 2024; 17:e13788. [PMID: 38561908 PMCID: PMC10985223 DOI: 10.1111/cts.13788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Predicting the subcutaneous (SC) pharmacokinetics (PK) of antibodies in humans is challenging, with clinical data currently being the only reliable data source for modeling SC absorption and bioavailability. Recombinant human hyaluronidase PH20 (rHuPH20) is an enzyme that facilitates SC delivery of high-dose, high-volume therapeutics. Numerous monoclonal antibodies have been co-administered SC with rHuPH20 in a clinical setting, establishing an extensive PK database. The goal of this work is to demonstrate how aggregated clinical data can be leveraged in a universal modeling framework for characterizing SC antibody PK, resulting in parameterization that can be used in predictive simulations of new antibodies. Data for 10 individual antibodies co-administered SC with rHuPH20 were obtained from publicly available sources. PK modeling of each antibody was conducted using the same model structure, but uniquely parameterized. The model structure consisted of a two-compartment model to capture linear kinetics, plus a target-binding mechanism to accommodate nonlinear kinetics driven by antibody-target complex formation and elimination. The clinical PK profiles for all antibodies were accurately described using the universal modeling framework. The SC PK parameters of absorption and bioavailability were consistent across the range of antibody and target properties evaluated. SC administration with rHuPH20 yielded a 30% increase in absorption rate on average and similar or better bioavailability. These parameter values can serve as initial conditions for model-based PK predictions for new antibodies co-administered SC with rHuPH20 to enable evaluation of optimal SC dose and schedule regimens prior to and during clinical development.
Collapse
|
27
|
Ait-Oudhia S, Wang YM, Dosne AG, Roy A, Jin JY, Shen J, Kagan L, Musuamba FT, Zhang L, Kijima S, Gastonguay MR, Ouellet D. Challenging the Norm: A Multidisciplinary Perspective on Intravenous to Subcutaneous Bridging Strategies for Biologics. Clin Pharmacol Ther 2024; 115:412-421. [PMID: 38069528 DOI: 10.1002/cpt.3133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023]
Abstract
The transition from intravenous (i.v.) to subcutaneous (s.c.) administration of biologics is a critical strategy in drug development aimed at improving patient convenience, compliance, and therapeutic outcomes. Focusing on the increasing role of model-informed drug development (MIDD) in the acceleration of this transition, an in-depth overview of the essential clinical pharmacology, and regulatory considerations for successful i.v. to s.c. bridging for biologics after the i.v. formulation has been approved are presented. Considerations encompass multiple aspects beginning with adequate pharmacokinetic (PK) and pharmacodynamic (i.e., exposure-response) evaluations which play a vital role in establishing comparability between the i.v. and s.c. routes of administrations. Selected key recommendations and points to consider include: (i) PK characterization of the s.c. formulation, supported by the increasing preclinical understanding of the s.c. absorption, and robust PK study design and analyses in humans; (ii) a thorough characterization of the exposure-response profiles including important metrics of exposure for both efficacy and safety; (iii) comparability studies designed to meet regulatory considerations and support approval of the s.c. formulation, including noninferiority studies with PK and/or efficacy and safety as primary end points; and (iv) comprehensive safety package addressing assessments of immunogenicity and patients' safety profile with the new route of administration. Recommendations for successful bridging strategies are evolving and MIDD approaches have been used successfully to accelerate the transition to s.c. dosing, ultimately leading to improved patient experiences, adherence, and clinical outcomes.
Collapse
Affiliation(s)
| | - Yow-Ming Wang
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anne-Gaelle Dosne
- Janssen Research & Development, LLC, Beerse, Belgium
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Amit Roy
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jin Y Jin
- Genentech Inc., South San Francisco, California, USA
| | - Jun Shen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Leonid Kagan
- Department of Pharmaceutics and Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Flora T Musuamba
- Belgian Federal Agency for Medicines and Health Products, Brussels, Belgium
- NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Lucia Zhang
- Health Canada, Biologic and Radiopharmaceutical Drugs Directorate, Ottawa, Ontario, Canada
| | - Shinichi Kijima
- Pharmaceuticals and Medical Devices Agency (PMDA), Tokyo, Japan
| | | | - Daniele Ouellet
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| |
Collapse
|
28
|
Hu G, Bonanno D, Su Y, Zhao X, Krishnamachari Y, Forrest W, Persak S, Givand J, Mannes D, Olbinado M, Wagner M, Grünzweig C, Novak V. Unraveling Pre-filled Syringe Needle Clogging: Exploring a Fresh Outlook Through Innovative Techniques. Pharm Res 2024; 41:547-556. [PMID: 38326531 DOI: 10.1007/s11095-024-03673-7] [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: 12/19/2023] [Accepted: 01/28/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVE This study aimed to investigate the movement of liquid in the needle region of staked-in-needle pre-filled syringes using neutron imaging and synchrotron X-ray tomography. The objective was to gain insights into the dynamics of liquid presence and understand the factors contributing to needle clogging. METHODS Staked-in-needle pre-filled syringes were examined using neutron radiography and synchrotron X-ray phase-contrast computed tomography. Neutron radiography provided a 2D visualization of liquid presence in the needle, while synchrotron X-ray tomography offered high-resolution 3D imaging to study detailed morphological features of the liquid. RESULTS Neutron radiography revealed liquid presence in the needle region for as-received samples and after temperature and pressure cycling. Pressure cycling had a more pronounced effect on liquid formation. Synchrotron X-ray tomography confirmed the presence of liquid and revealed various morphologies, including droplets of different sizes, liquid segments blocking sections of the needle, and a thin layer covering the needle wall. Liquid presence was also observed between the steel needle and the glass barrel. CONCLUSIONS The combination of neutron imaging and synchrotron X-ray tomography provided valuable insights into the dynamics of liquid movement in staked-in-needle pre-filled syringes. Temperature and pressure cycling were found to contribute to additional liquid formation, with pressure changes playing a significant role. The detailed morphological analysis enhanced the understanding of microstructural arrangements within the needle. This research contributes to addressing the issue of needle clogging and can guide the development of strategies to improve pre-filled syringe performance.
Collapse
Affiliation(s)
- Guangli Hu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA.
| | - Daniel Bonanno
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Yongchao Su
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA.
| | - Xi Zhao
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Yogita Krishnamachari
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - William Forrest
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Steven Persak
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Jeffrey Givand
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - David Mannes
- ANAXAM, PARK INNOVAARE: deliveryLAB, CH-5234, Villigen, Switzerland
- Paul Scherrer Institute (PSI), Forschungsstrasse 111, 5232, Villigen, Switzerland
| | - Margie Olbinado
- ANAXAM, PARK INNOVAARE: deliveryLAB, CH-5234, Villigen, Switzerland
- Paul Scherrer Institute (PSI), Forschungsstrasse 111, 5232, Villigen, Switzerland
| | - Matthias Wagner
- ANAXAM, PARK INNOVAARE: deliveryLAB, CH-5234, Villigen, Switzerland
| | | | - Vladimir Novak
- ANAXAM, PARK INNOVAARE: deliveryLAB, CH-5234, Villigen, Switzerland
| |
Collapse
|
29
|
Davis JD, Bravo Padros M, Conrado DJ, Ganguly S, Guan X, Hassan HE, Hazra A, Irvin SC, Jayachandran P, Kosloski MP, Lin KJ, Mukherjee K, Paccaly A, Papachristos A, Partridge MA, Prabhu S, Visich J, Welf ES, Xu X, Zhao A, Zhu M. Subcutaneous Administration of Monoclonal Antibodies: Pharmacology, Delivery, Immunogenicity, and Learnings From Applications to Clinical Development. Clin Pharmacol Ther 2024; 115:422-439. [PMID: 38093583 DOI: 10.1002/cpt.3150] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/06/2023] [Indexed: 01/11/2024]
Abstract
Subcutaneous (s.c.) administration of monoclonal antibodies (mAbs) can reduce treatment burden for patients and healthcare systems compared with intravenous (i.v.) infusion through shorter administration times, made possible by convenient, patient-centric devices. A deeper understanding of clinical pharmacology principles related to efficacy and safety of s.c.-administered mAbs over the past decade has streamlined s.c. product development. This review presents learnings from key constituents of the s.c. mAb development pathway, including pharmacology, administration variables, immunogenicity, and delivery devices. Restricted mAb transportation through the hypodermis explains their incomplete absorption at a relatively slow rate (pharmacokinetic (PK)) and may impact mAb-cellular interactions and/or onset and magnitude of physiological responses (pharmacodynamic). Injection volumes, formulation, rate and site of injection, and needle attributes may affect PKs and the occurrence/severity of adverse events like injection-site reactions or pain, with important consequences for treatment adherence. A review of immunogenicity data for numerous compounds reveals that incidence of anti-drug antibodies (ADAs) is generally comparable across i.v. and s.c. routes, and complementary factors including response magnitude (ADA titer), persistence over time, and neutralizing antibody presence are needed to assess clinical impact. Finally, four case studies showcase how s.c. biologics have been clinically developed: (i) by implementation of i.v./s.c. bridging strategies to streamline PD-1/PD-L1 inhibitor development, (ii) through co-development with i.v. presentations for anti-severe acute respiratory syndrome-coronavirus 2 antibodies to support rapid deployment of both formulations, (iii) as the lead route for bispecific T cell engagers (BTCEs) to mitigate BTCE-mediated cytokine release syndrome, and (iv) for pediatric patients in the case of dupilumab.
Collapse
Affiliation(s)
- John D Davis
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | | | - Samit Ganguly
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Xiaowen Guan
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Hazem E Hassan
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Anasuya Hazra
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Susan C Irvin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | | | - Kuan-Ju Lin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | - Anne Paccaly
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | | | - Saileta Prabhu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | - Erik S Welf
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Xiaoying Xu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - An Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Min Zhu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| |
Collapse
|
30
|
Rodriguez-Otero P, van de Donk NWCJ, Pillarisetti K, Cornax I, Vishwamitra D, Gray K, Hilder B, Tolbert J, Renaud T, Masterson T, Heuck C, Kane C, Verona R, Moreau P, Bahlis N, Chari A. GPRC5D as a novel target for the treatment of multiple myeloma: a narrative review. Blood Cancer J 2024; 14:24. [PMID: 38307865 PMCID: PMC10837198 DOI: 10.1038/s41408-023-00966-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 02/04/2024] Open
Abstract
Multiple myeloma is a genetically complex and heterogenous malignancy with a 5-year survival rate of approximately 60%. Despite advances in therapy, patients experience cycles of remission and relapse, with each successive line of therapy associated with poorer outcomes; therefore, therapies with different mechanisms of action against new myeloma antigens are needed. G protein-coupled receptor class C group 5 member D (GPRC5D) has emerged as a novel therapeutic target for the treatment of multiple myeloma. We review the biology and target validation of GPRC5D, and clinical data from early phase trials of GPRC5D-targeting bispecific antibodies, talquetamab and forimtamig, and chimeric antigen receptor T cell (CAR-T) therapies, MCARH109, OriCAR-017, and BMS-986393. In addition to adverse events (AEs) associated with T-cell-redirection therapies irrespective of target, a consistent pattern of dermatologic and oral AEs has been reported across several trials of GPRC5D-targeting bispecific antibodies, as well as rare cerebellar events with CAR-T therapy. Additional studies are needed to understand the underlying mechanisms involved in the development of skin- and oral-related toxicities. We review the strategies that have been used to manage these GPRC5D-related toxicities. Preliminary efficacy data showed overall response rates for GPRC5D-targeting T-cell-redirecting therapies were ≥64%; most responders achieved a very good partial response or better. Pharmacokinetics/pharmacodynamics showed that these therapies led to cytokine release and T-cell activation. In conclusion, results from early phase trials of GPRC5D-targeting T-cell-redirecting agents have shown promising efficacy and manageable safety profiles, including lower infection rates compared with B-cell maturation antigen- and Fc receptor-like protein 5-targeting bispecific antibodies. Further clinical trials, including those investigating GPRC5D-targeting T-cell-redirecting agents in combination with other anti-myeloma therapies and with different treatment modalities, may help to elucidate the future optimal treatment regimen and sequence for patients with multiple myeloma and improve survival outcomes. Video Summary.
Collapse
Affiliation(s)
| | - Niels W C J van de Donk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | - Brandi Hilder
- Janssen Research & Development, Spring House, PA, USA
| | | | | | | | | | - Colleen Kane
- Janssen Research & Development, Spring House, PA, USA
| | - Raluca Verona
- Janssen Research & Development, Spring House, PA, USA
| | | | - Nizar Bahlis
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ajai Chari
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
31
|
Zhong X, Liu Y, Ardekani AM. A compartment model for subcutaneous injection of monoclonal antibodies. Int J Pharm 2024; 650:123687. [PMID: 38103705 DOI: 10.1016/j.ijpharm.2023.123687] [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: 05/27/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
Despite the growing popularity of subcutaneous (SC) administration for monoclonal antibodies (mAbs), there remains a limited understanding of the significance of mAb transport rate constants within the interstitial space and the lymphatic system on their pharmacokinetics. To bridge this knowledge gap, we introduce a compartmental model for subcutaneously administered mAbs. Our model differentiates FcRn-expressing cells across various sites, and the model predictions agree with experimental data from both human and rat studies. Our findings indicate that the time to reach the maximum mAb concentration in the plasma, denoted by Tmax, displays a weak positive correlation with mAb half-life and a negligible correlation with bioavailability. In contrast, the half-life of mAbs exhibits a strong positive correlation with bioavailability. Moreover, the rate of mAb transport from lymph to plasma significantly affects the mAb half-life. Increasing the transport rates of mAbs from the injection site to the lymph or from lymph to plasma enhances bioavailability. These insights, combined with our compartmental model, contribute to a deeper understanding of the pharmacokinetics of subcutaneously administered mAbs.
Collapse
Affiliation(s)
- Xiaoxu Zhong
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States
| | - Yikai Liu
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States.
| |
Collapse
|
32
|
Dahmana N, Destruel PL, Facchetti S, Braun V, Lebouc V, Marin Z, Patel S, Schwach G. Reversible protein complexes as a promising avenue for the development of high concentration formulations of biologics. Int J Pharm 2023; 648:123616. [PMID: 37977291 DOI: 10.1016/j.ijpharm.2023.123616] [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: 09/05/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
High concentration formulations have become an important pre-requisite in the development of biological drugs, particularly in the case of subcutaneous administration where limited injection volume negatively affects the administered dose. In this study, we propose to develop high concentration formulations of biologics using a reversible protein-polyelectrolyte complex (RPC) approach. First, the versatility of RPC was assessed using different complexing agents and formats of therapeutic proteins, to define the optimal conditions for complexation and dissociation of the complex. The stability of the protein was investigated before and after complexation, as well as upon a 4-week storage period at various temperatures. Subsequently, two approaches were selected to develop high concentration RPC formulations: first, using up-concentrated RPC suspensions in aqueous buffers, and second, by generating spray-dried RPC and further resuspension in non-aqueous solvents. Results showed that the RPC concept is applicable to a wide range of therapeutic protein formats and the complexation-dissociation process did not affect the stability of the proteins. High concentration formulations up to 200 mg/mL could be achieved by up-concentrating RPC suspensions in aqueous buffers and RPC suspensions in non-aqueous solvents were concentrated up to 250 mg/mL. Although optimization is needed, our data suggests that RPC may be a promising avenue to achieve high concentration formulations of biologics for subcutaneous administration.
Collapse
Affiliation(s)
- Naoual Dahmana
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Pierre-Louis Destruel
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland.
| | - Samantha Facchetti
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Vanessa Braun
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Vanessa Lebouc
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Zana Marin
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Sulabh Patel
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Gregoire Schwach
- Pharmaceutical Development & Supplies, PTD Biologics Europe, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| |
Collapse
|
33
|
Stielow M, Witczyńska A, Kubryń N, Fijałkowski Ł, Nowaczyk J, Nowaczyk A. The Bioavailability of Drugs-The Current State of Knowledge. Molecules 2023; 28:8038. [PMID: 38138529 PMCID: PMC10745386 DOI: 10.3390/molecules28248038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Drug bioavailability is a crucial aspect of pharmacology, affecting the effectiveness of drug therapy. Understanding how drugs are absorbed, distributed, metabolized, and eliminated in patients' bodies is essential to ensure proper and safe treatment. This publication aims to highlight the relevance of drug bioavailability research and its importance in therapy. In addition to biochemical activity, bioavailability also plays a critical role in achieving the desired therapeutic effects. This may seem obvious, but it is worth noting that a drug can only produce the expected effect if the proper level of concentration can be achieved at the desired point in a patient's body. Given the differences between patients, drug dosages, and administration forms, understanding and controlling bioavailability has become a priority in pharmacology. This publication discusses the basic concepts of bioavailability and the factors affecting it. We also looked at various methods of assessing bioavailability, both in the laboratory and in the clinic. Notably, the introduction of new technologies and tools in this field is vital to achieve advances in drug bioavailability research. This publication also discusses cases of drugs with poorly described bioavailability, providing a deeper understanding of the complex challenges they pose to medical researchers and practitioners. Simultaneously, the article focuses on the perspectives and trends that may shape the future of research regarding bioavailability, which is crucial to the development of modern pharmacology and drug therapy. In this context, the publication offers an essential, meaningful contribution toward understanding and highlighting bioavailability's role in reliable patient treatment. The text also identifies areas that require further research and exploration.
Collapse
Affiliation(s)
| | - Adrianna Witczyńska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 Jurasza Street, 85-089 Bydgoszcz, Poland; (A.W.); (N.K.); (Ł.F.)
| | - Natalia Kubryń
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 Jurasza Street, 85-089 Bydgoszcz, Poland; (A.W.); (N.K.); (Ł.F.)
| | - Łukasz Fijałkowski
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 Jurasza Street, 85-089 Bydgoszcz, Poland; (A.W.); (N.K.); (Ł.F.)
| | - Jacek Nowaczyk
- Department of Physical Chemistry and Physicochemistry of Polymers, Faculty of Chemistry, Nicolaus Copernicus University, 7 Gagarina Street, 87-100 Toruń, Poland;
| | - Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 Jurasza Street, 85-089 Bydgoszcz, Poland; (A.W.); (N.K.); (Ł.F.)
| |
Collapse
|
34
|
Barlek MH, Gillis DC, Egner SA, Maragos SL, Karver MR, Stupp SI, Tsihlis ND, Kibbe MR. Systemic peptide amphiphile nanofiber delivery following subcutaneous injection. Biomaterials 2023; 303:122401. [PMID: 38006645 DOI: 10.1016/j.biomaterials.2023.122401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Peptide amphiphile (PA) nanofibers have been shown to target and deliver drugs when administered via an intravenous (IV) injection. Subcutaneous administration can broaden the applicability of PA nanofibers in the medical field. The ability of PA nanofibers to be absorbed into systemic circulation after subcutaneous administration was investigated. Four PA molecules with different amino acid sequences were designed to understand the effect of nanofiber cohesion and charge on uptake. Solution small-angle X-ray scattering confirmed nanostructure morphology and provided characteristic lengths for co-assemblies. Circular dichroism and solution wide-angle X-ray scattering confirmed PA secondary structure and molecular order. PAs were co-assembled in a 95 %:5 % molar ratio of unlabeled PA to fluorescently labeled PA. Male and female Sprague Dawley rats were injected in the nape of the neck with PA co-assemblies. In vivo normalized abdominal fluorescence was measured 1-72 h after injection. PA nanofibers with a negative charge and low internal order showed the highest amount of systemic absorption at 1, 6, and 24 h. At 24 h after injection, white blood cell count decreased and glucose was elevated. Glucose began to decrease at 48 h. These data indicate that PA nanofibers can be absorbed into the systemic circulation after subcutaneous injection.
Collapse
Affiliation(s)
- Mark H Barlek
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA
| | - David C Gillis
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA
| | - Simon A Egner
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Sophia L Maragos
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA
| | - Mark R Karver
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, 60611, USA; Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Samuel I Stupp
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA; Departments of Chemistry and Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, 60611, USA; Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Nick D Tsihlis
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - Melina R Kibbe
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904, USA.
| |
Collapse
|
35
|
Wang H, Hu T, Leng Y, de Lucio M, Gomez H. MPET 2: a multi-network poroelastic and transport theory for predicting absorption of monoclonal antibodies delivered by subcutaneous injection. Drug Deliv 2023; 30:2163003. [PMID: 36625437 PMCID: PMC9851243 DOI: 10.1080/10717544.2022.2163003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Subcutaneous injection of monoclonal antibodies (mAbs) has attracted much attention in the pharmaceutical industry. During the injection, the drug is delivered into the tissue producing strong fluid flow and tissue deformation. While data indicate that the drug is initially uptaken by the lymphatic system due to the large size of mAbs, many of the critical absorption processes that occur at the injection site remain poorly understood. Here, we propose the MPET2 approach, a multi-network poroelastic and transport model to predict the absorption of mAbs during and after subcutaneous injection. Our model is based on physical principles of tissue biomechanics and fluid dynamics. The subcutaneous tissue is modeled as a mixture of three compartments, i.e., interstitial tissue, blood vessels, and lymphatic vessels, with each compartment modeled as a porous medium. The proposed biomechanical model describes tissue deformation, fluid flow in each compartment, the fluid exchanges between compartments, the absorption of mAbs in blood vessels and lymphatic vessels, as well as the transport of mAbs in each compartment. We used our model to perform a high-fidelity simulation of an injection of mAbs in subcutaneous tissue and evaluated the long-term drug absorption. Our model results show good agreement with experimental data in depot clearance tests.
Collapse
Affiliation(s)
- Hao Wang
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA,CONTACT Hao Wang School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Tianyi Hu
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Yu Leng
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Mario de Lucio
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Hector Gomez
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
36
|
Torres JE, Meng F, Bhattacharya S, Buno KP, Ahmadzadegan A, Madduri S, Babiak PM, Vlachos PP, Solorio L, Yeo Y, Liu JC. Interpenetrating Networks of Collagen and Hyaluronic Acid That Serve as In Vitro Tissue Models for Assessing Macromolecular Transport. Biomacromolecules 2023; 24:4718-4730. [PMID: 37651737 DOI: 10.1021/acs.biomac.3c00448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
High-fidelity preclinical in vitro tissue models can reduce the failure rate of drugs entering clinical trials. Collagen and hyaluronic acid (HA) are major components of the extracellular matrix of many native tissues and affect therapeutic macromolecule diffusion and recovery through tissues. Although collagen and HA are commonly used in tissue engineering, the physical and mechanical properties of these materials are variable and depend highly on processing conditions. In this study, HA was chemically modified and crosslinked via hydrazone bonds to form interpenetrating networks of crosslinked HA (HAX) with collagen (Col). These networks enabled a wide range of mechanical properties, including stiffness and swellability, and microstructures, such as pore morphology and size, that can better recapitulate diverse tissues. We utilized these interpenetrating ColHAX hydrogels as in vitro tissue models to examine macromolecular transport and recovery for early-stage drug screening. Hydrogel formulations with varying collagen and HAX concentrations imparted different gel properties based on the ratio of collagen to HAX. These gels were stable and swelled up to 170% of their original mass, and the storage moduli of the ColHAX gels increased over an order of magnitude by increasing collagen and HA concentration. Interestingly, when HAX concentration was constant and collagen concentration increased, both the pore size and spatial colocalization of collagen and HA increased. HA in the system dominated the ζ-potentials of the gels. The hydrogel and macromolecule properties impacted the mass transport and recovery of lysozyme, β-lactoglobulin, and bovine serum albumin (BSA) from the ColHAX gels─large molecules were largely impacted by mesh size, whereas small molecules were influenced primarily by electrostatic forces. Overall, the tunable properties demonstrated by the ColHAX hydrogels can be used to mimic different tissues for early-stage assays to understand drug transport and its relationship to matrix properties.
Collapse
Affiliation(s)
- Jessica E Torres
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Fanfei Meng
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sayantan Bhattacharya
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kevin P Buno
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Adib Ahmadzadegan
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sathvik Madduri
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Pavlos P Vlachos
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
37
|
Khadria A, Paavola CD, Maslov K, Brown-Augsburger PL, Grealish PF, Lozano E, Blankenship RL, Cao R, Shi J, Beals JM, Oladipupo SS, Wang LV. Photoacoustic imaging of the dynamics of a dye-labeled IgG4 monoclonal antibody in subcutaneous tissue reveals a transient decrease in murine blood oxygenation under anesthesia. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:116002. [PMID: 38078154 PMCID: PMC10704085 DOI: 10.1117/1.jbo.28.11.116002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023]
Abstract
Significance Over 100 monoclonal antibodies have been approved by the U.S. Food and Drug Administration (FDA) for clinical use; however, a paucity of knowledge exists regarding the injection site behavior of these formulated therapeutics, particularly the effect of antibody, formulation, and tissue at the injection site. A deeper understanding of antibody behavior at the injection site, especially on blood oxygenation through imaging, will help design improved versions of the therapeutics for a wide range of diseases. Aim The aim of this research is to understand the dynamics of monoclonal antibodies at the injection site as well as how the antibody itself affects the functional characteristics of the injection site [e.g., blood oxygen saturation (sO 2 )]. Approach We employed triple-wavelength equipped functional photoacoustic imaging to study the dynamics of dye-labeled and unlabeled monoclonal antibodies at the site of injection in a mouse ear. We injected a near-infrared dye-labeled (and unlabeled) human IgG4 isotype control antibody into the subcutaneous space in mouse ears to analyze the injection site dynamics and quantify molecular movement, as well as its effect on local hemodynamics. Results We performed pharmacokinetic studies of the antibody in different regions of the mouse body to show that dye labeling does not alter the pharmacokinetic characteristics of the antibody and that mouse ear is a viable model for these initial studies. We explored the movement of the antibody in the interstitial space to show that the bolus area grows by ∼ 300 % over 24 h. We discovered that injection of the antibody transiently reduces the local sO 2 levels in mice after prolonged anesthesia without affecting the total hemoglobin content and oxygen extraction fraction. Conclusions This finding on local oxygen saturation opens a new avenue of study on the functional effects of monoclonal antibody injections. We also show the suitability of the mouse ear model to study antibody dynamics through high-resolution imaging techniques. We quantified the movement of antibodies at the injection site caused by the interstitial fluid, which could be helpful for designing antibodies with tailored absorption speeds in the future.
Collapse
Affiliation(s)
- Anjul Khadria
- California Institute of Technology, Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Pasadena, California, United States
| | - Chad D. Paavola
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, Indiana, United States
| | - Konstantin Maslov
- California Institute of Technology, Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Pasadena, California, United States
| | - Patricia L. Brown-Augsburger
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, Indiana, United States
| | - Patrick F. Grealish
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, Indiana, United States
| | - Emmanuel Lozano
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, Indiana, United States
| | - Ross L. Blankenship
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, Indiana, United States
| | - Rui Cao
- California Institute of Technology, Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Pasadena, California, United States
| | - Junhui Shi
- California Institute of Technology, Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Pasadena, California, United States
| | - John M. Beals
- Eli Lilly and Company, Lilly Biotechnology Center, Lilly Research Laboratories, San Diego, California, United States
| | - Sunday S. Oladipupo
- Eli Lilly and Company, Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, Indiana, United States
| | - Lihong V. Wang
- California Institute of Technology, Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Pasadena, California, United States
- California Institute of Technology, Caltech Optical Imaging Laboratory, Department of Electrical Engineering, Pasadena, California, United States
| |
Collapse
|
38
|
Zeng Y, Naik S, Tran T, Wuthrich P, Muni N, Mahoney RP. Preclinical Pharmacokinetic Study on Caffeine as an Excipient for Monoclonal Antibody Formulations. J Pharm Sci 2023; 112:2933-2937. [PMID: 37517525 DOI: 10.1016/j.xphs.2023.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
Caffeine is a novel excipient that effectively reduces viscosity of high concentration mAb formulations intended for subcutaneous (SQ) delivery. Two preclinical studies were conducted in rats to evaluate pharmacokinetic (PK) parameters of caffeine as well as its effects on the PK profile of a model mAb, namely ipilimumab. Results show that SQ absorption and elimination of caffeine was rapid, with the average Tmax of 0.4 h and T1/2 of 1.6 h, administered with or without ipilimumab. Furthermore, caffeine did not affect ipilimumab SQ PK profiles. Independent of caffeine concentration, ipilimumab serum T1/2 was between 2 and 3 days, Tmax was between 3 and 4 days and SQ bioavailability was about 64%. In addition, SQ injection of caffeine at different dose levels showed no irritation at the injection site or adverse effects. Results from the current PK studies warrant further development of caffeine as a viscosity reducing excipient for mAb SQ formulations.
Collapse
Affiliation(s)
- Yuhong Zeng
- Comera Life Sciences, Inc., 12 Gill Street Suite 4650, Woburn, MA 01801, USA.
| | - Subhashchandra Naik
- Comera Life Sciences, Inc., 12 Gill Street Suite 4650, Woburn, MA 01801, USA
| | - Timothy Tran
- Comera Life Sciences, Inc., 12 Gill Street Suite 4650, Woburn, MA 01801, USA
| | - Philip Wuthrich
- Comera Life Sciences, Inc., 12 Gill Street Suite 4650, Woburn, MA 01801, USA
| | - Neal Muni
- Comera Life Sciences, Inc., 12 Gill Street Suite 4650, Woburn, MA 01801, USA
| | - Robert P Mahoney
- Comera Life Sciences, Inc., 12 Gill Street Suite 4650, Woburn, MA 01801, USA
| |
Collapse
|
39
|
Chandran Suja V, Qi QM, Halloran K, Zhang J, Shaha S, Prakash S, Kumbhojkar N, Deslandes A, Huille S, Gokarn YR, Mitragotri S. A biomimetic chip to assess subcutaneous bioavailability of monoclonal antibodies in humans. PNAS NEXUS 2023; 2:pgad317. [PMID: 37901442 PMCID: PMC10612570 DOI: 10.1093/pnasnexus/pgad317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023]
Abstract
Subcutaneous (subQ) injection is a common route for delivering biotherapeutics, wherein pharmacokinetics is largely influenced by drug transport in a complex subQ tissue microenvironment. The selection of good drug candidates with beneficial pharmacokinetics for subQ injections is currently limited by a lack of reliable testing models. To address this limitation, we report here a Subcutaneous Co-Culture Tissue-on-a-chip for Injection Simulation (SubCuTIS). SubCuTIS possesses a 3D coculture tissue architecture, and it allows facile quantitative determination of relevant scale independent drug transport rate constants. SubCuTIS captures key in vivo physiological characteristics of the subQ tissues, and it differentiates the transport behavior of various chemically distinct molecules. We supplemented the transport measurements with theoretical modeling, which identified subtle differences in the local absorption rate constants of seven clinically available mAbs. Accounting for first-order proteolytic catabolism, we established a mathematical framework to assess clinical bioavailability using the local absorption rate constants obtained from SubCuTIS. Taken together, the technology described here broadens the applicability of organs-on-chips as a standardized and easy-to-use device for quantitative analysis of subQ drug transport.
Collapse
Affiliation(s)
- Vineeth Chandran Suja
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Qin M Qi
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | | | | | - Suyog Shaha
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Supriya Prakash
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Ninad Kumbhojkar
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | | | - Sylvain Huille
- Sanofi R&D, Impasse Des Ateliers, Vitry-sur-Seine 94400 France
| | | | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| |
Collapse
|
40
|
Hauser SL, Kappos L, Bar-Or A, Wiendl H, Paling D, Williams M, Gold R, Chan A, Milo R, Das Gupta A, Karlsson G, Sullivan R, Graham G, Merschhemke M, Häring DA, Vermersch P. The Development of Ofatumumab, a Fully Human Anti-CD20 Monoclonal Antibody for Practical Use in Relapsing Multiple Sclerosis Treatment. Neurol Ther 2023; 12:1491-1515. [PMID: 37450172 PMCID: PMC10444716 DOI: 10.1007/s40120-023-00518-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023] Open
Abstract
The importance of B cells in multiple sclerosis (MS) has been demonstrated through the advent of B-cell-depleting anti-CD20 antibody therapies. Ofatumumab is the first fully human anti-CD20 monoclonal antibody (mAb) developed and tested for subcutaneous (SC) self-administration at monthly doses of 20 mg, and has been approved in the US, UK, EU, and other regions and countries worldwide for the treatment of relapsing MS. The development goal of ofatumumab was to obtain a highly efficacious anti-CD20 therapy, with a safety and tolerability profile that allows for self-administration by MS patients at home and a positive benefit-risk balance for use in the broad relapsing MS population. This development goal was enabled by the unique binding site, higher affinity to B cells, and higher potency of ofatumumab compared to previous anti-CD20 mAbs; these properties of ofatumumab facilitate rapid B-cell depletion and maintenance with a low dose at a low injection volume (20 mg/0.4 ml). The high potency in turn enables the selective targeting of B cells that reside in the lymphatic system via subcutaneous (SC) administration. Through a comprehensive dose-finding program in two phase 2 studies (one intravenous and one SC) and model simulations, it was found that safety and tolerability can be further improved, and the risk of systemic injection-related reactions (IRRs) minimized, by avoiding doses ≥ 30 mg, and by reaching initial and rapid B-cell depletion via stepwise weekly administration of ofatumumab at Weeks 0, 1, and 2 (instead of a single high dose). Once near-complete B-cell depletion is reached, it can be maintained by monthly doses of 20 mg/0.4 ml. Indeed, in phase 3 trials (ASCLEPIOS I/II), rapid and sustained near-complete B-cell depletion (largely independent of body weight, race and other factors) was observed with this dosing regimen, which resulted in superior efficacy of ofatumumab versus teriflunomide on relapse rates, disability worsening, neuronal injury (serum neurofilament light chain), and imaging outcomes. Likely due to its fully human nature, ofatumumab has a low immunogenic risk profile-only 2 of 914 patients receiving ofatumumab in ASCLEPIOS I/II developed anti-drug antibodies-and this may also underlie the infrequent IRRs (20% with ofatumumab vs. 15% with the placebo injection in the teriflunomide arm) that were mostly (99.8%) mild to moderate in severity. The overall rates of infections and serious infections in patients treated with ofatumumab were similar to those in patients treated with teriflunomide (51.6% vs. 52.7% and 2.5% vs. 1.8%, respectively). The benefit-risk profile of ofatumumab was favorable compared to teriflunomide in the broad RMS population, and also in the predefined subgroups of both recently diagnosed and/or treatment-naïve patients, as well as previously disease-modifying therapy-treated patients. Interim data from the ongoing extension study (ALITHIOS) have shown that long-term treatment with ofatumumab up to 4 years is well-tolerated in RMS patients, with no new safety risks identified. In parallel to the phase 3 trials in which SC administration was carried out with a pre-filled syringe, an autoinjector pen for more convenient self-administration of the ofatumumab 20 mg dose was developed and is available for use in clinical practice.
Collapse
Affiliation(s)
- Stephen L Hauser
- UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Ludwig Kappos
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) and MS Center, and Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - David Paling
- Sheffield Institute of Translational Neuroscience, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Mitzi Williams
- Joi Life Wellness Multiple Sclerosis Neurology Center, Atlanta, GA, USA
| | - Ralf Gold
- Department of Neurology, St Josef-Hospital/Ruhr-University Bochum, Bochum, Germany
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ron Milo
- Department of Neurology, Barzilai Medical Center, Ashkelon/Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | | | | - Patrick Vermersch
- Univ. Lille, INSERM U1172 LilNCog, CHU Lille, FHU Precise, 59000, Lille, France
| |
Collapse
|
41
|
Yokoyama M, Suzuki E, Oitate M, Watanabe N. A Quantitative Prediction Method for the Human Pharmacokinetics of Fc-Fusion Proteins. Eur J Drug Metab Pharmacokinet 2023; 48:541-552. [PMID: 37530974 DOI: 10.1007/s13318-023-00845-5] [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: 07/04/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND AND OBJECTIVE Fc fusion is an effective strategy for extending the half-lives of therapeutic proteins. This study aimed to evaluate the applicability of a human pharmacokinetics prediction method for Fc-fusion proteins by extending on reported methods for monoclonal antibodies (mAbs). METHODS To predict human pharmacokinetic profiles following intravenous (IV) dosing, the pharmacokinetic data for 11 Fc-fusion proteins in monkeys were analysed by two approaches: a species-invariant time method with a range of allometric exponents in clearance (CL, 0.7-1.0) and a two-compartment model reported for mAbs. The pharmacokinetic profiles following subcutaneous (SC) dosing were predicted by simple dose normalisation from monkeys or using the geometric means of the absorption rate constant (Ka) and bioavailability (BA) for mAbs or Fc-fusion proteins in humans and compared. RESULTS In the case of IV administration, the area under the curve could be predicted for more than 85% of Fc-fusion proteins within a twofold difference from the observed value using the species-invariant time method (scaling exponent for CL, 0.95). For SC dosing, incorporating the geometric means of absorption parameters for both mAbs (BA 68.2%, Ka 0.287 day-1) and Fc-fusion proteins (BA 63.0%, Ka 0.209 day-1) in humans provided better accuracy than simple normalisation from monkeys. CONCLUSION We have successfully predicted the human pharmacokinetic profiles of Fc-fusion proteins for both IV and SC administration within twofold of the observed value from monkey pharmacokinetic data by extending on reported methods for mAbs. This method will facilitate drug discovery and development of Fc-fusion proteins.
Collapse
Affiliation(s)
- Miki Yokoyama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan.
| | - Eiko Suzuki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Masataka Oitate
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Nobuaki Watanabe
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| |
Collapse
|
42
|
Sree VD, Toaquiza-Tubon JD, Payne J, Solorio L, Tepole AB. Damage and Fracture Mechanics of Porcine Subcutaneous Tissue Under Tensile Loading. Ann Biomed Eng 2023; 51:2056-2069. [PMID: 37233856 DOI: 10.1007/s10439-023-03233-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/04/2023] [Indexed: 05/27/2023]
Abstract
Subcutaneous injection, which is a preferred delivery method for many drugs, causes deformation, damage, and fracture of the subcutaneous tissue. Yet, experimental data and constitutive modeling of these dissipation mechanisms in subcutaneous tissue remain limited. Here we show that subcutaneous tissue from the belly and breast anatomical regions in the swine show nonlinear stress-strain response with the characteristic J-shaped behavior of collagenous tissue. Additionally, subcutaneous tissue experiences damage, defined as a decrease in the strain energy capacity, as a function of the previously experienced maximum deformation. The elastic and damage response of the tissue are accurately described by a microstructure-driven constitutive model that relies on the convolution of a neo-Hookean material of individual fibers with a fiber orientation distribution and a fiber recruitment distribution. The model fit revealed that subcutaneous tissue can be treated as initially isotropic, and that changes in the fiber recruitment distribution with loading are enough to explain the dissipation of energy due to damage. When tested until failure, subcutaneous tissue that has undergone damage fails at the same peak stress as virgin samples, but at a much larger stretch, overall increasing the tissue toughness. Together with a finite element implementation, these data and constitutive model may enable improved drug delivery strategies and other applications for which subcutaneous tissue biomechanics are relevant.
Collapse
Affiliation(s)
- Vivek D Sree
- School of Mechanical Engineering, Purdue University, West Lafayette, USA
| | | | - Jordanna Payne
- School of Mechanical Engineering, Purdue University, West Lafayette, USA
| | - Luis Solorio
- School of Mechanical Engineering, Purdue University, West Lafayette, USA
| | | |
Collapse
|
43
|
M Morris N, A Blee J, Hauert S. Global parameter optimisation and sensitivity analysis of antivenom pharmacokinetics and pharmacodynamics. Toxicon 2023; 232:107206. [PMID: 37356552 DOI: 10.1016/j.toxicon.2023.107206] [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: 03/14/2023] [Revised: 06/05/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
In recent years it has become possible to design snakebite antivenoms with diverse pharmacokinetic properties. Owing to the pharmacokinetic variability of venoms, the choice of antivenom scaffold may influence a treatment's neutralisation coverage. Computation offers a useful medium through which to assess the pharmacokinetics and pharmacodynamics of envenomation-treatment systems, as antivenoms with identical neutralising capacities can be simulated. In this study, we simulate envenomation and treatment with a variety of antivenoms, to define the properties of effective antivenoms. Systemic envenomation and treatment were described using a two-compartment pharmacokinetic model. Treatment of Naja sumatrana and Cryptelytrops purpureomaculatus envenomation was simulated with a set of 200,000 theoretical antivenoms across 10 treatment time delays. These two venoms are well-characterised and have differing pharmacokinetic properties. The theoretical antivenom set varied across molecular weight, dose, kon, koff, and valency. The best and worst treatments were identified using an area under the curve metric, and a global sensitivity analysis was performed to quantify the influence of the input parameters on treatment outcome. The simulations show that scaffolds of diverse molecular formats can be effective. Molecular weight and valency have a negligible direct impact on treatment outcome, however low molecular weight scaffolds offer more flexibility across the other design parameters, particularly when treatment is delayed. The simulations show kon to primarily mediate treatment efficacy, with rates above 105 M-1s-1 required for the most effective treatments. koff has the greatest impact on the performance of less effective scaffolds. While the same scaffold preferences for improved treatment are seen for both model snakes, the parameter bounds for C. purpureomaculatus envenomation are more constrained. This paper establishes a computational framework for the optimisation of antivenom design.
Collapse
Affiliation(s)
- Natalie M Morris
- Department of Engineering Mathematics, Ada Lovelace Building, University of Bristol, University Walk, Bristol, BS8 1TW, UK.
| | - Johanna A Blee
- Department of Engineering Mathematics, Ada Lovelace Building, University of Bristol, University Walk, Bristol, BS8 1TW, UK.
| | - Sabine Hauert
- Department of Engineering Mathematics, Ada Lovelace Building, University of Bristol, University Walk, Bristol, BS8 1TW, UK.
| |
Collapse
|
44
|
Bittner B, Sánchez-Félix M, Lee D, Koynov A, Horvath J, Schumacher F, Matoori S. Drug delivery breakthrough technologies - A perspective on clinical and societal impact. J Control Release 2023; 360:335-343. [PMID: 37364797 DOI: 10.1016/j.jconrel.2023.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
The way a drug molecule is administered has always had a profound impact on people requiring medical interventions - from vaccine development to cancer therapeutics. In the Controlled Release Society Fall Symposium 2022, a trans-institutional group of scientists from industry, academia, and non-governmental organizations discussed what a breakthrough in the field of drug delivery constitutes. On the basis of these discussions, we classified drug delivery breakthrough technologies into three categories. In category 1, drug delivery systems enable treatment for new molecular entities per se, for instance by overcoming biological barriers. In category 2, drug delivery systems optimize efficacy and/or safety of an existing drug, for instance by directing distribution to their target tissue, by replacing toxic excipients, or by changing the dosing reqimen. In category 3, drug delivery systems improve global access by fostering use in low-resource settings, for instance by facilitating drug administration outside of a controlled health care institutional setting. We recognize that certain breakthroughs can be classified in more than one category. It was concluded that in order to create a true breakthrough technology, multidisciplinary collaboration is mandated to move from pure technical inventions to true innovations addressing key current and emerging unmet health care needs.
Collapse
Affiliation(s)
- Beate Bittner
- Global Product Strategy, Product Optimization, Grenzacher Strasse 124, 4070 Basel, Switzerland.
| | - Manuel Sánchez-Félix
- Novartis Institutes for BioMedical Research, 700 Main Street, Cambridge, MA 02139, USA
| | - Dennis Lee
- Bill & Melinda Gates Foundation, Seattle, WA 98119, United States
| | - Athanas Koynov
- Pharmaceutical Sciences, Merck & Co., Inc., Rahway, NJ 07033, United States
| | - Joshua Horvath
- Device and Packaging Development, Genentech, Inc., South San Francisco, CA, United States
| | - Felix Schumacher
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Simon Matoori
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
45
|
Dos Santos ACF, Ahmadzadegan A, Ximenes E, Vlachos P, Ardekani A, Kapur S, Corvari V, Ladisch MR. Concentration-dependent diffusion of unlabeled protein within an in vitro hyaluronic acid matrix. Biotechnol Bioeng 2023. [PMID: 37466320 DOI: 10.1002/bit.28505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/16/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
Diffusion and movement of subcutaneously injected biologics and high-concentration immunoglobulin G (IgG) therapeutics away from the injection site and through the subcutaneous (SC) tissue may be concentration dependent. This possibility was confirmed by in situ measurement of diffusion coefficients of unlabeled bovine IgG in phosphate-buffered saline within an in vitro hyaluronic acid matrix that represents the SC electrostatic environment. Diffusion decreased from 2.67 to 0.05 × 10-7 cm2 /s when IgG concentration increased from 25 to 73 mg/mL. The results demonstrated that in situ detection of unlabeled proteins within an in vitro SC environment provides another useful tool for the preclinical characterization of injectable biologics.
Collapse
Affiliation(s)
- Antonio C F Dos Santos
- Department of Agricultural and Biological Engineering, Laboratory of Renewable Resources Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Adib Ahmadzadegan
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Eduardo Ximenes
- Department of Agricultural and Biological Engineering, Laboratory of Renewable Resources Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Pavlos Vlachos
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Arezoo Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Shiven Kapur
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Vince Corvari
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Michael R Ladisch
- Department of Agricultural and Biological Engineering, Laboratory of Renewable Resources Engineering, Purdue University, West Lafayette, Indiana, USA
- Eli Lilly, Indianapolis, Indiana, USA
| |
Collapse
|
46
|
von Richter O, O'Reilly T, Guerrieri D, Fan J, Fey C, Schussler S, Furlan F, Lemke L. GP2017-HCF, a high concentration formulation, demonstrates similar pharmacokinetics, immunogenicity and safety to GP2017, an approved adalimumab biosimilar. Expert Opin Biol Ther 2023; 23:749-758. [PMID: 36039657 DOI: 10.1080/14712598.2022.2117546] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/23/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND GP2017 is an adalimumab biosimilar. The objective of this study is to compare the pharmacokinetics (PK) of GP2017 in its approved formulation and GP2017-high concentration formulation (HCF) in a randomized, double-blind, two-arm PK bridging study. RESEARCH DESIGN AND METHODS Healthy male subjects received a single 40 mg subcutaneous injection of either GP2017-HCF (n = 162) or GP2017 (n = 168). PK, safety, and immunogenicity were assessed over 72 days post-injection. RESULTS The 90% confidence intervals [CIs] of geometric mean ratios between GP2017-HCF and GP2017 for Cmax, AUC0-inf, AUC0-360 and AUC0-last were within the pre-defined margin of 0.80 to 1.25; thus, PK comparability between GP2017-HCF and GP2017 was demonstrated. Subgroup analysis of PK comparability by anti-drug antibody (ADA) subpopulation showed that the 90% CIs of geometric mean ratios between GP2017-HCF and GP2017 for Cmax, AUC0-inf, AUC0-360 and AUC0-last were within the margin of 0.80 to 1.25 in ADA-positive and ADA-negative subjects. The proportions of subjects with positive ADA responses and with neutralizing antibodies were comparable between the GP2017-HCF and GP2017 groups. GP2017-HCF and GP2017 were well tolerated, and there were no reports of deaths or other serious adverse events. CONCLUSION Results show PK comparability between GP2017-HCF and GP2017 and comparable safety and tolerability.
Collapse
Affiliation(s)
- Oliver von Richter
- Clinical Development Biopharmaceuticals, Hexal AG (a Sandoz company), Holzkirchen, Germany
| | | | - Davide Guerrieri
- Clinical Development Biopharmaceuticals, Hexal AG (a Sandoz company), Holzkirchen, Germany
| | - Jamie Fan
- BioPharma Clinical Development, Sandoz Inc, Princeton, NJ, USA
| | - Constanze Fey
- Clinical Development Biopharmaceuticals, Hexal AG (a Sandoz company), Holzkirchen, Germany
| | | | - Fabricio Furlan
- Global Medical Affairs, Biopharmaceuticals, Hexal AG (a Sandoz company), Holzkirchen, Germany
| | - Lena Lemke
- Clinical Development Biopharmaceuticals, Hexal AG (a Sandoz company), Holzkirchen, Germany
| |
Collapse
|
47
|
Wang S, Liaw A, Chen YM, Su Y, Skomski D. Convolutional Neural Networks Enable Highly Accurate and Automated Subvisible Particulate Classification of Biopharmaceuticals. Pharm Res 2023; 40:1447-1457. [PMID: 36471026 DOI: 10.1007/s11095-022-03438-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Quantification of subvisible particles, which are generally defined as those ranging in size from 2 to 100 µm, is important as critical characteristics for biopharmaceutical formulation development. Micro Flow Imaging (MFI) provides quantifiable morphological parameters to study both the size and type of subvisible particles, including proteinaceous particles as well as non-proteinaceous features incl. silicone oil droplets, air bubble droplets, etc., thus enabling quantitative and categorical particle attribute reporting for quality control. However, limitations in routine MFI image analysis can hinder accurate subvisible particle classification. In this work, we custom-built a subvisible particle-aware Convolutional Neural Network, SVNet, which has a very small computational footprint, and achieves comparable performance to prior state-of-art image classification models. SVNet significantly improves upon current standard operating procedures for subvisible particulate assessments as confirmed by thorough real-world validation studies.
Collapse
Affiliation(s)
- Shubing Wang
- Department of Biometrics Research, Merck & Co. Inc., 126 East Lincoln Avenue, Rahway, New Jersey, 07065, USA.
| | - Andy Liaw
- Department of Biometrics Research, Merck & Co. Inc., 126 East Lincoln Avenue, Rahway, New Jersey, 07065, USA
| | - Yue-Ming Chen
- Department of Biometrics Research, Merck & Co. Inc., 126 East Lincoln Avenue, Rahway, New Jersey, 07065, USA
| | - Yongchao Su
- Analytical Research and Development, Merck & Co. Inc., 126 East Lincoln Avenue, Rahway, New Jersey, 07065, USA
| | - Daniel Skomski
- Analytical Research and Development, Merck & Co. Inc., 126 East Lincoln Avenue, Rahway, New Jersey, 07065, USA.
| |
Collapse
|
48
|
Zou P. Predicting Human Bioavailability of Subcutaneously Administered Monoclonal Antibodies Using Non-human Primate Linear Clearance and Antibody Isoelectric Point. AAPS J 2023; 25:53. [PMID: 37225958 DOI: 10.1208/s12248-023-00818-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
The prediction of bioavailability is one of the major barriers in the clinical translation of subcutaneously (SC) administered therapeutic monoclonal antibodies (mAbs) due to the lack of reliable in vitro and preclinical in vivo predictive models. Recently, multiple linear regression (MLR) models were developed to predict human SC bioavailability of mAbs using human linear clearance (CL) and isoelectric point (pI) of the whole antibody or Fv regions as independent variables. Unfortunately, these models cannot be applied to mAbs at the preclinical development stage because human CLs of these mAbs are unknown. In this study, we predicted human SC bioavailability of mAbs using preclinical data only by two approaches. In the first approach, allometric scaling was used to predict human linear CL from non-human primate (NHP) linear CL. The predicted human CL and the pI of the whole antibody or Fv regions were then incorporated into two previously published MLR models to predict the human bioavailability of 61 mAbs. In the second approach, two MLR models were developed using NHP linear CL and the pI of whole antibody or Fv regions of 41 mAbs in a training set. The two models were validated using an independent test dataset containing 20 mAbs. The four MLR models generated 77-85% of predictions within 0.8- to 1.2-fold deviations from observed human bioavailability. Overall, this study demonstrated that human SC bioavailability of mAbs at the preclinical stage could be predicted using NHP CL and pI of mAbs.
Collapse
Affiliation(s)
- Peng Zou
- Quantitative Clinical Pharmacology, Daiichi Sankyo, Inc., 211 Mt. Airy Road, Basking Ridge, New Jersey, 07920, USA.
| |
Collapse
|
49
|
Abd-Ellah HS, Mudududdla R, Carter GP, Baell JB. Novel Perspectives on the Design and Development of a Long-Acting Subcutaneous Raltegravir Injection for Treatment of HIV-In Vitro and In Vivo Evaluation. Pharmaceutics 2023; 15:pharmaceutics15051530. [PMID: 37242770 DOI: 10.3390/pharmaceutics15051530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Antiretrovirals (ARVs) are a highly effective therapy for treatment and prevention of HIV infection, when administered as prescribed. However, adherence to lifelong ARV regimens poses a considerable challenge and places HIV patients at risk. Long-acting ARV injections may improve patient adherence as well as maintaining long-term continuous drug exposure, resulting in improved pharmacodynamics. In the present work, we explored the aminoalkoxycarbonyloxymethyl (amino-AOCOM) ether prodrug concept as a potential approach to long-acting ARV injections. As a proof of concept, we synthesised model compounds containing the 4-carboxy-2-methyl Tokyo Green (CTG) fluorophore and assessed their stability under pH and temperature conditions that mimic those found in the subcutaneous (SC) tissue. Among them, probe 21 displayed very slow fluorophore release under SC-like conditions (98% of the fluorophore released over 15 d). Compound 25, a prodrug of the ARV agent raltegravir (RAL), was subsequently prepared and evaluated using the same conditions. This compound showed an excellent in vitro release profile, with a half-life (t½) of 19.3 d and 82% of RAL released over 45 d. In mice, 25 extended the half-life of unmodified RAL by 4.2-fold (t½ = 3.18 h), providing initial proof of concept of the ability of amino-AOCOM prodrugs to extend drug lifetimes in vivo. Although this effect was not as pronounced as seen in vitro-presumably due to enzymatic degradation and rapid clearance of the prodrug in vivo-the present results nevertheless pave the way for development of more metabolically stable prodrugs, to facilitate long-acting delivery of ARVs.
Collapse
Affiliation(s)
- Heba S Abd-Ellah
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Ramesh Mudududdla
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia
| | - Glen P Carter
- Microbiology and Immunology Department, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3001, Australia
| | - Jonathan B Baell
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, China
| |
Collapse
|
50
|
Chang HP, Le HK, Shah DK. Pharmacokinetics and Pharmacodynamics of Antibody-Drug Conjugates Administered via Subcutaneous and Intratumoral Routes. Pharmaceutics 2023; 15:pharmaceutics15041132. [PMID: 37111619 PMCID: PMC10142912 DOI: 10.3390/pharmaceutics15041132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
We hypothesize that different routes of administration may lead to altered pharmacokinetics/pharmacodynamics (PK/PD) behavior of antibody-drug conjugates (ADCs) and may help to improve their therapeutic index. To evaluate this hypothesis, here we performed PK/PD evaluation for an ADC administered via subcutaneous (SC) and intratumoral (IT) routes. Trastuzumab-vc-MMAE was used as the model ADC, and NCI-N87 tumor-bearing xenografts were used as the animal model. The PK of multiple ADC analytes in plasma and tumors, and the in vivo efficacy of ADC, after IV, SC, and IT administration were evaluated. A semi-mechanistic PK/PD model was developed to characterize all the PK/PD data simultaneously. In addition, local toxicity of SC-administered ADC was investigated in immunocompetent and immunodeficient mice. Intratumoral administration was found to significantly increase tumor exposure and anti-tumor activity of ADC. The PK/PD model suggested that the IT route may provide the same efficacy as the IV route at an increased dosing interval and reduced dose level. SC administration of ADC led to local toxicity and reduced efficacy, suggesting difficulty in switching from IV to SC route for some ADCs. As such, this manuscript provides unprecedented insight into the PK/PD behavior of ADCs after IT and SC administration and paves the way for clinical evaluation of these routes.
Collapse
Affiliation(s)
- Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| | - Huyen Khanh Le
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| |
Collapse
|