1
|
Albach FN, Geier C, Keicher C, Posch MG, Schreiber SJ, Grütz G, Akyüz L, Luo X, Le-Halpere A, Truffinet P, Wagner F. Phase 1 Trials of Gatralimab, a Next-Generation Humanized Anti-CD52 Monoclonal Antibody, in Participants with Progressive Multiple Sclerosis. Neurol Ther 2024; 13:1607-1625. [PMID: 39251561 PMCID: PMC11541066 DOI: 10.1007/s40120-024-00659-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
INTRODUCTION Lymphocyte depletion via anti-CD52 monoclonal antibody (mAb) therapy is an effective treatment strategy for relapsing-remitting multiple sclerosis (MS) but is associated with infusion/injection-associated reactions (IARs) and autoimmune-related adverse events (AEs). Gatralimab is a next-generation humanized anti-CD52 mAb. METHODS Two first-in-human trials were conducted in participants with progressive MS to assess the pharmacodynamics, pharmacokinetics, and safety of gatralimab administered via subcutaneous (SC) and intravenous (IV) routes, and to determine the effect of different comedication regimes on IARs to SC gatralimab. A Phase 1 trial (NCT02282826) included double-blind, placebo-controlled sequential ascending single IV (1, 3.5, and 12 mg) and SC (12, 36, and 60 mg) dose groups. A Phase 1b trial (NCT02977533) involved five groups who received SC gatralimab (36, 48, or 60 mg) and different comedications. A long-term safety (LTS) study (NCT02313285) examined safety and pharmacodynamics over 4 years. RESULTS Gatralimab produced depletion of lymphocytes (dose-dependently) and CD4+ regulatory T cells, with partial repopulation to normal values by approximately 12 months. Peak serum gatralimab concentrations followed dose-proportionality and were delayed by 6.0-7.5 days following SC administration. Treatment-emergent AEs, including IARs, were reported for most participants but were generally of mild or moderate severity, and treatment-emergent serious AEs were mostly MS-related. Methylprednisolone and antihistamine comedications were associated with reduced incidence of fevers and skin and subcutaneous tissue AEs, respectively. During the LTS study, one participant (3.0%) experienced an autoimmune-related AE (Basedow's disease), and subsequently died from pulmonary sepsis deemed unrelated to gatralimab by the investigator. CONCLUSIONS These data show that gatralimab achieves the desired pharmacodynamic effect of lymphocyte depletion followed by repopulation, and has an acceptable safety profile, including low risk of non-MS autoimmunity. Although gatralimab is no longer in development for MS, insights from these trials may inform the development of comedication regimes of future anti-CD52 mAbs and subcutaneous formulations of other lymphocyte-depleting mAbs. TRIAL REGISTRATION NCT02282826, NCT02977533, NCT02313285.
Collapse
Affiliation(s)
| | | | | | | | | | - Gerald Grütz
- BIH Centre for Regenerative Therapies (BCRT), Immunocheck-Biomarker Unit, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Levent Akyüz
- BIH Centre for Regenerative Therapies (BCRT), Immunocheck-Biomarker Unit, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | - Frank Wagner
- Charité Research Organisation GmbH, Berlin, Germany
| |
Collapse
|
2
|
Koenitz L, Crean A, Vucen S. Pharmacokinetic differences between subcutaneous injection and intradermal microneedle delivery of protein therapeutics. Eur J Pharm Biopharm 2024; 204:114517. [PMID: 39349073 DOI: 10.1016/j.ejpb.2024.114517] [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: 07/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
Protein therapeutics are essential in the treatment of various diseases, but most of them require parenteral administration. Since intravenous and subcutaneous injections are associated with discomfort and pain, other routes have been investigated including intradermal microneedle delivery. Microneedles are shorter than hypodermic needles and therefore minimize contact with pain receptors in deeper skin layers. But the differences in anatomical and physiological characteristics of dermis and subcutis can potentially result in varying protein penetration through the skin, absorption, and metabolism. This review summarizes pharmacokinetic studies that compare the administration of protein therapeutics by subcutaneous injections and different types of microneedles intradermally including hollow, dissolvable, coated, and hydrogel-forming microneedles. Across animal and human studies, hollow microneedle delivery resulted in quicker and higher peak plasma levels of proteins and comparable bioavailability to subcutaneous injections potentially due to the extensive network of lymphatic and blood vessels in the dermis. In case of dissolvable and coated microneedles, drug release kinetics depend on component materials. The dissolution of polymer excipients can slow the release and permeation of protein therapeutics at the administration site and thereby delay absorption. The understanding of drug penetration through different skin layers, its absorption into blood capillaries or lymphatics, and dermal metabolism remains limited. Additionally, the effects of these processes on the differences in pharmacokinetic profiles of proteins following intradermal microneedle administration are not well understood. Greater insights are required for the development of the next generation of intradermal microneedle biotherapeutics.
Collapse
Affiliation(s)
- Laura Koenitz
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland.
| | - Abina Crean
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| | - Sonja Vucen
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
3
|
Xu C, Xin K, Kosloski MP, Butler A, Goulaouic H, Nivens MC, Kanamaluru V. Pharmacokinetics of Subcutaneous Itepekimab Injection With an Autoinjector Device and Prefilled Syringe in Healthy Participants. Clin Pharmacol Drug Dev 2024; 13:1181-1188. [PMID: 39308293 DOI: 10.1002/cpdd.1466] [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: 03/02/2024] [Accepted: 08/13/2024] [Indexed: 11/06/2024]
Abstract
Itepekimab, a monoclonal antibody against interleukin-33, has demonstrated clinical utility in previous studies in patients with asthma and chronic obstructive pulmonary disease. An autoinjector (AI) has been developed for administering itepekimab to facilitate further development. This study compared pharmacokinetics of single 300-mg itepekimab subcutaneous administration via an AI versus a prefilled syringe (PFS). Of 90 healthy volunteers enrolled in this Phase 1, parallel-design, randomized study and stratified by body weight (50 to <70 kg, ≥70 to <80 kg, ≥80 to 100 kg) and injection site (abdomen, thigh, or arm), 84 completed the study. Systemic exposure of itepekimab was similar for both groups. Point estimates for geometric mean ratios of pharmacokinetic parameters for AI versus PFS groups were 1.01 for maximum serum concentration, 1.06 for area under the serum concentration-time curve to the last quantifiable concentration, and 1.04 for area under the serum concentration-time curve extrapolated to infinity. The exposure was similar for both devices in each body weight and injection site subgroup. Overall, systemic exposure of 300-mg single-dose itepekimab in healthy participants was comparable when administered subcutaneously via an AI device and PFS, with an acceptable safety profile in both device groups.
Collapse
|
4
|
Janssen JC, van Dijk B, Hoeijmakers LL, Grünhagen DJ, Bramer WM, Verhoef C, de Gruijl TD, Blank CU, van der Veldt AAM. Local administration of immunotherapy for patients with skin cancer: A systematic review. Cancer Treat Rev 2024; 131:102848. [PMID: 39486396 DOI: 10.1016/j.ctrv.2024.102848] [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: 09/18/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024]
Abstract
Since the introduction of immune checkpoint inhibitors (ICIs) targeting PD-1 and CTLA-4 receptors, survival has improved significantly for patients with irresectable and metastatic skin cancer, including cutaneous squamous cell cancer and melanoma. However, systemic administration of these drugs is associated with immune related adverse events (irAEs), which can be severe, irreversible and even fatal. To reduce the risk of irAEs associated with systemic exposure to immunotherapeutic drugs, local administration of low doses could be considered. This systematic review provides an overview of early phase clinical trials with drugs that are currently under investigation for intratumoral administration in patients with melanoma and non-melanoma skin cancer.
Collapse
Affiliation(s)
- J C Janssen
- Department of Medical Oncology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Department of Surgical Oncology and Gastro Intestinal Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - B van Dijk
- Department of Medical Oncology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - L L Hoeijmakers
- Department of Medical Oncology, Antoni van Leeuwenhoek - Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D J Grünhagen
- Department of Surgical Oncology and Gastro Intestinal Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - W M Bramer
- Medical Library, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - C Verhoef
- Department of Surgical Oncology and Gastro Intestinal Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - T D de Gruijl
- Department of Immunology, Amsterdam UMC, University Medical Center, Amsterdam, the Netherlands
| | - C U Blank
- Department of Medical Oncology, Antoni van Leeuwenhoek - Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - A A M van der Veldt
- Department of Medical Oncology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
5
|
Parlow J, Rodler A, Gråsjö J, Sjögren H, Hansson P. FRAP analysis of peptide diffusion in extracellular matrix mimetic hydrogels as an in vitro model for subcutaneous injection. Int J Pharm 2024; 664:124628. [PMID: 39179009 DOI: 10.1016/j.ijpharm.2024.124628] [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: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Subcutaneous (SC) injection is a common route of administration for drug compounds with poor oral bioavailability. However, bioavailability is often variable and incomplete, and there is as yet no standard accepted medium for simulation of the human SC environment. In this work we evaluate a FRAP based method for quantitative determination of local self-diffusion coefficients within extracellular matrix (ECM) mimetic hydrogels, potentially useful as in vitro models for drug transport in the ECM after SC injection. Gels were made consisting of either agarose, cross-linked collagen (COL) and hyaluronic acid (HA) or cross-linked HA. The diffusivities of uncharged FITC-dextran (FD4), the highly charged poly-lysine (PLK20) and poly-glutamic acid (PLE20) as well as the GLP-1 analogue exenatide were determined within the gels using FRAP. The diffusion coefficients in uncharged agarose gels were in the range of free diffusion in PBS. The diffusivity of cationic PLK20 in gels containing anionic HA was substantially decreased due to strong electrostatic interactions. Peptide aggregation could be observed as immobile fractions in experiments with exenatide. We conclude that the FRAP method provides useful information of peptides' interactions and transport properties in hydrogel networks, giving insight into the mechanisms affecting absorption of drug compounds after subcutaneous injection.
Collapse
Affiliation(s)
- Julia Parlow
- Department of Medicinal Chemistry, Uppsala University, SE-75123 Uppsala, Sweden
| | - Agnes Rodler
- Department of Medicinal Chemistry, Uppsala University, SE-75123 Uppsala, Sweden
| | - Johan Gråsjö
- Department of Medicinal Chemistry, Uppsala University, SE-75123 Uppsala, Sweden
| | - Helen Sjögren
- Ferring Pharmaceuticals A/S, Amager Strandvej 405, DK-2770 Kastrup, Denmark
| | - Per Hansson
- Department of Medicinal Chemistry, Uppsala University, SE-75123 Uppsala, Sweden.
| |
Collapse
|
6
|
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] [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
|
7
|
Zur Y, Katchkovsky S, Itzhar A, Abramovitch-Dahan CV, Stepensky D, Papo N, Levaot N. Preventing osteoporotic bone loss in mice by promoting balanced bone remodeling through M-CSF RGD, a dual antagonist to c-FMS and αvβ3 receptors. Int J Biol Macromol 2024; 282:136821. [PMID: 39447795 DOI: 10.1016/j.ijbiomac.2024.136821] [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: 06/23/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Osteoporosis is a common, age-related disease caused by imbalanced bone remodeling. Current treatments either shut down bone resorption or robustly stimulate bone formation. Here, we describe a novel compound that inhibits osteoclast activity without causing apparent disruptions to bone formation by targeting both c-FMS (i.e., osteoclast differentiation) and αvβ3 integrin (i.e., osteoclastic bone resorption) receptors. We show that human serum albumin (HSA)-conjugated M-CSFRGD protein (M-CSFRGD-HSA) effectively inhibits the activity of both receptors, with a three-fold higher serum half-life compared to the unconjugated M-CSFRGD. We then treated ovariectomized mice with different doses of M-CSFRGD-HSA, alendronate, or a monospecific control protein. The bispecific M-CSFRGD-HSA was superior to a monospecific control in alleviating bone loss and reducing osteoclast distribution and function. M-CSFRGD-HSA and alendronate effectively prevented ovariectomy-induced bone loss, but M-CSFRGD-HSA had a milder inhibitory effect on osteoclast distribution and activity. Moreover, alendronate halted bone formation, while M-CSFRGD-HSA-treated mice showed an increased level of serum amino-terminal propeptide of type I collagen, a bone formation marker. Our data indicate that the mild reduction in osteoclast activity facilitated by the bispecific M-CSFRGD-HSA allows the maintenance of certain levels of bone formation and may be superior to treatments that induce osteoclast depletion.
Collapse
Affiliation(s)
- Yuval Zur
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Svetlana Katchkovsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Amit Itzhar
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Chen-Viki Abramovitch-Dahan
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - David Stepensky
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | - Noam Levaot
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
8
|
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] [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
|
9
|
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
|
10
|
Jacquot G, Lopez Navarro P, Grange C, Boudali L, Harlepp S, Pivot X, Detappe A. Landscape of Subcutaneous Administration Strategies for Monoclonal Antibodies in Oncology. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406604. [PMID: 39165046 DOI: 10.1002/adma.202406604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/15/2024] [Indexed: 08/22/2024]
Abstract
In recent decades, subcutaneous (SC) administration of monoclonal antibodies (mAbs) has emerged as a promising alternative to intravenous delivery in oncology, offering comparable therapeutic efficacy while addressing patient preferences. This perspective article provides an in-depth analysis of the technological landscape surrounding SC mAb administration in oncology. It outlines various technologies under evaluation across developmental stages, spanning from preclinical investigations to the integration of established methodologies in clinical practice. Additionally, this perspective article explores emerging trends and prospective trajectories, shedding light on the evolving landscape of SC mAb administration. Furthermore, it emphasizes key checkpoints related to quality attributes essential for optimizing mAb delivery via the SC route. This review serves as a valuable resource for researchers, clinicians, and healthcare policymakers, offering insights into the advancement of SC mAb administration in oncology and its implications for patient care.
Collapse
Affiliation(s)
- Guillaume Jacquot
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Pedro Lopez Navarro
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Coralie Grange
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Lotfi Boudali
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Sébastien Harlepp
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
- Equipe de Synthèse Pour l'Analyse, Institut Pluridisciplinaire Hubert Curien (IPHC), UMR 7178 CNRS/University of Strasbourg, Strasbourg, Cedex 2, 67087, France
| |
Collapse
|
11
|
Nolan RP, Kang DW, Maneval DC, Knowles SP, LaBarre MJ, Printz MA. The safety of recombinant human hyaluronidase PH20 in nonclinical models: An overview of toxicology, pharmacology, and impact of anti-PH20 antibodies. J Control Release 2024; 374:369-383. [PMID: 39089505 DOI: 10.1016/j.jconrel.2024.07.062] [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: 04/26/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
Hyaluronan (HA) is a glycosaminoglycan that forms a gel-like barrier in the subcutaneous (SC) space, limiting bulk fluid flow and the dispersion of SC-administered therapeutics. Recombinant human hyaluronidase PH20 (rHuPH20) facilitates the rapid delivery of co-administered therapeutics by depolymerizing HA in the SC space. Administration of rHuPH20 can induce the formation of anti-rHuPH20 antibodies, or anti-drug antibodies (ADAs), with the potential to bind endogenous PH20 hyaluronidase in the adult testes and epididymis. Using a variety of relevant animal models and multiple dose regimens of rHuPH20 across the full spectrum of animal development, we demonstrated that rHuPH20 administration resulted in the formation of ADAs. Although these ADAs can bind both the recombinant rHuPH20 enzyme and recombinant versions of animal model-specific hyaluronidases, they had no impact on fertility parameters (as measured by sperm concentration and motility, litter size, and litter viability) or fetal development. We present the result of our nonclinical studies in order of the developmental lifecycle, beginning with adults. Toxicology studies that extend beyond the standard package are also presented. These studies demonstrate the favorable safety profile of rHuPH20 and ADAs in nonclinical models. Additionally, we identified substantial safety margins for clinically relevant doses of rHuPH20.
Collapse
|
12
|
Bryniarski MA, Tuhin MTH, Shomin CD, Nasrollahi F, Ko EC, Soto M, Chung K, Poon-Andersen C, Primack R, Wong D, Ojeda E, Chung J, Cook KD, Conner KP. Utility of Cellular Measurements of Non-Specific Endocytosis to Assess the Target-Independent Clearance of Monoclonal Antibodies. J Pharm Sci 2024; 113:3100-3111. [PMID: 39009346 DOI: 10.1016/j.xphs.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Past studies have demonstrated higher clearance for monoclonal antibodies possessing increased rates of non-specific endocytosis. However, this metric is oftentimes evaluated indirectly using biophysical techniques or cell surface binding studies that may not provide insight into the specific rates of cellular turnover. Furthermore, few examples evaluating non-specific endocytosis have been reported for a therapeutic antibody that reached clinical assessment. In the current report, we evaluated a therapeutic human immunoglobulin G2 monoclonal antibody targeted against the interleukin-4 receptor alpha chain (IL-4Rα) that exhibited elevated target independent clearance in previous Phase 1 and 2 studies. We confirmed high non-specific clearance of the anti-IL-4Rα antibody as compared to a reference antibody during pharmacokinetic assessments in wild type mice where target-mediated disposition was absent. We then developed a cell-based method capable of measuring cellular protein endocytosis and demonstrated the anti-IL-4Rα antibody exhibited marked non-specific uptake relative to the reference compound. Antibody homology modeling identified the anti-IL-4Rα antibody possessed positive charge patches whose removal via targeted mutations substantially reduced its non-specific endocytosis. We then expanded the scope of the study by evaluating panels of both preclinical and clinically relevant monoclonal antibodies and demonstrate those with the highest rates of non-specific uptake in vitro exhibited elevated target independent clearance, low subcutaneous bioavailability, or both. Our results support the observation that high non-specific endocytosis is a negative attribute in monoclonal antibody development and demonstrate the utility of a generic cell-based screen as a quantitative tool to measure non-specific endocytosis of protein therapeutics at the single-cell level.
Collapse
Affiliation(s)
- Mark A Bryniarski
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA.
| | - Md Tariqul Haque Tuhin
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Carolyn D Shomin
- Department of Biologics, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Fatemeh Nasrollahi
- Process Development; Pre-Pivotal Drug Product Technologies, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Eunkyung Clare Ko
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Marcus Soto
- Pharmacokinetics & Drug Metabolism, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Kyu Chung
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Carrie Poon-Andersen
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Ronya Primack
- Pharmacokinetics & Drug Metabolism, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Diana Wong
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Esperanza Ojeda
- Pharmacokinetics & Drug Metabolism, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - John Chung
- Process Development; Pre-Pivotal Drug Product Technologies, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Kevin D Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Kip P Conner
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA.
| |
Collapse
|
13
|
Ozdemir Kutahya Z, Gokbulut C, Aslan Akyol B, Yavuz O, Sen F, Piner Benli P. The effects of breed and routes of administration on the plasma pharmacokinetics and faecal excretion of robenacoxib in goats. Vet J 2024; 307:106200. [PMID: 39095012 DOI: 10.1016/j.tvjl.2024.106200] [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: 04/09/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Robenacoxib (RX) is a non-steroidal anti-inflammatory drug (NSAID) of the coxib class. This study aimed to evaluate the plasma dispositions and faecal excretion profiles of RX in Alpine and Saanen goats following oral and subcutaneous routes. Two different goat breeds were allocated into two treatment groups concerning the breed. RX was administered subcutaneously to animals at a dose of 4 mg/kg b.w. Following a one-week washout period, RX was administered by oral route to the same animals at the same dose. Heparinized blood samples were collected from all animals before drug administration (0 h) and subsequently up to 24 h. Faecal samples were collected at various times between 8 h and 36 h. The concentrations of RX in plasma and faeces were determined by HPLC. The plasma half-life (T1/2λz) of RX in Saanen goats (1.21 h) was significantly longer (P < 0.017) than in Alpine goats (0.90 h) after subcutaneous administration. In both goat breeds, statistical differences were observed between subcutaneous and oral administration of RX for T1/2λz, Tlast, Cmax, AUC0-∞, and MRT0-∞. Faecal Cmax and Tmax parameters following oral administrations were 0.92 µg/g and 0.85 µg/g at 30 h and at 24 h in Alpine and Saanen goats, respectively. The difference in plasma protein ratio between Alpine and Saanen goats may have affected the T1/2λz of the drug. NSAIDs are among the drug groups frequently detected in aquatic and terrestrial ecosystems around the world and there are data on the effects of NSAID residues on wildlife and aquatic species. Therefore, revealing the excretion of NSAIDs, which are frequently used in the veterinary field, in faeces and urine should be considered for ecological sustainability.
Collapse
Affiliation(s)
- Zeynep Ozdemir Kutahya
- Cukurova University, Faculty of Ceyhan Veterinary Medicine, Department of Veterinary Pharmacology and Toxicology, Adana, Türkiye.
| | - Cengiz Gokbulut
- Balikesir University, Faculty of Medicine, Department of Medical Pharmacology, Balikesir, Türkiye; Institute of Health Sciences, Department of Veterinary Pharmacology and Toxicology, Balikesir, Türkiye
| | - Busra Aslan Akyol
- Balikesir University, Institute of Health Sciences, Department of Veterinary Pharmacology and Toxicology, CoHE 100/2000 Scholarship Holder, Balikesir, Türkiye
| | - Oguzhan Yavuz
- Cukurova University, Faculty of Ceyhan Veterinary Medicine, Department of Veterinary Pharmacology and Toxicology, Adana, Türkiye
| | - Fazilet Sen
- Balikesir University, Faculty of Medicine, Department of Medical Pharmacology, Balikesir, Türkiye
| | - Petek Piner Benli
- Cukurova University, Faculty of Ceyhan Veterinary Medicine, Department of Veterinary Pharmacology and Toxicology, Adana, Türkiye
| |
Collapse
|
14
|
Ayoun Alsoud R, Le Moan N, Holten-Andersen L, Knudsen T, Lennernäs H, Simonsson USH. Model-Based Interspecies Scaling for Predicting Human Pharmacokinetics of CB 4332, a Complement Factor I Protein. J Pharm Sci 2024; 113:2895-2903. [PMID: 38945365 DOI: 10.1016/j.xphs.2024.06.022] [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: 03/04/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Interspecies scaling of the pharmacokinetics (PK) of CB 4332, a 150 kDa recombinant complement factor I protein, was performed using traditional and model-based approaches to inform first-in-human dose selection. Plasma concentration versus time data from four preclinical PK studies of single intravenous and subcutaneous (SC) CB 4332 dosing in mice, rats and nonhuman primates (NHPs) were modeled simultaneously using naive pooling including allometric scaling. The human-equivalent dose was calculated using the preclinical no observed adverse effect level (NOAEL) as part of the dose-by-factor approach. Pharmacokinetic modeling of CB 4332 revealed species-specific differences in the elimination, which was accounted for by including an additional rat-specific clearance. Signs of anti-drug antibodies (ADA) formation in all rats and some NHPs were observed. Consequently, an additional ADA-induced clearance parameter was estimated including the time of onset. The traditional dose-by-factor approach calculated a maximum recommended starting SC dose of 0.9 mg/kg once weekly, which was predicted it to result in a trough steady-state concentration lower than the determined efficacy target range for CB 4332 in humans. Model simulations predicted the efficacy target range to be reached using 5 mg/kg once weekly SC dosing.
Collapse
Affiliation(s)
- Rami Ayoun Alsoud
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Tom Knudsen
- Catalyst Biosciences, South San Francisco, California, USA
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
15
|
Li C, Zhong X, Rahimi E, Ardekani AM. A multi-scale numerical study of monoclonal antibodies uptake by initial lymphatics after subcutaneous injection. Int J Pharm 2024; 661:124419. [PMID: 38972522 DOI: 10.1016/j.ijpharm.2024.124419] [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: 02/13/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024]
Abstract
This paper studies the transport of monoclonal antibodies through skin tissue and initial lymphatics, which impacts the pharmacokinetics of monoclonal antibodies. Our model integrates a macroscale representation of the entire skin tissue with a mesoscale model that focuses on the papillary dermis layer. Our results indicate that it takes hours for the drugs to disperse from the injection site to the papillary dermis before entering the initial lymphatics. Additionally, we observe an inhomogeneous drug distribution in the interstitial space of the papillary dermis, with higher drug concentrations near initial lymphatics and lower concentrations near blood capillaries. To validate our model, we compare our numerical simulation results with experimental data, finding a good alignment. Our parametric studies on the drug molecule properties and injection parameters suggest that a higher diffusion coefficient increases the transport and uptake rate while binding slows down these processes. Furthermore, shallower injection depths lead to faster lymphatic uptake, whereas the size of the injection plume has a minor effect on the uptake rate. These findings advance our understanding of drug transport and lymphatic absorption after subcutaneous injection, offering valuable insights for optimizing drug delivery strategies and the design of biotherapeutics.
Collapse
Affiliation(s)
- Chenji Li
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States
| | - Xiaoxu Zhong
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States
| | - Ehsan Rahimi
- 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
|
16
|
de Lucio M, Leng Y, Wang H, Vlachos PP, Gomez H. Modeling drug transport and absorption in subcutaneous injection of monoclonal antibodies: Impact of tissue deformation, devices, and physiology. Int J Pharm 2024; 661:124446. [PMID: 38996825 DOI: 10.1016/j.ijpharm.2024.124446] [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: 04/24/2024] [Revised: 06/21/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
The pharmaceutical industry has experienced a remarkable increase in the use of subcutaneous injection of monoclonal antibodies (mAbs), attributed mainly to its advantages in reducing healthcare-related costs and enhancing patient compliance. Despite this growth, there is a limited understanding of how tissue mechanics, physiological parameters, and different injection devices and techniques influence the transport and absorption of the drug. In this work, we propose a high-fidelity computational model to study drug transport and absorption during and after subcutaneous injection of mAbs. Our numerical model includes large-deformation mechanics, fluid flow, drug transport, and blood and lymphatic uptake. Through this computational framework, we analyze the tissue material responses, plume dynamics, and drug absorption. We analyze different devices, injection techniques, and physiological parameters such as BMI, flow rate, and injection depth. Finally, we compare our numerical results against the experimental data from the literature.
Collapse
Affiliation(s)
- Mario de Lucio
- 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, Los Alamos, NM 87544, USA
| | - Hao Wang
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| | - Pavlos P Vlachos
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| | - Hector Gomez
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| |
Collapse
|
17
|
Hakim MH, Brindise MC, Ahmadzadegan A, Buno KP, Dos Santos ACF, Cragg KR, Dou Z, Ladisch MR, Ardekani AM, Vlachos PP, Solorio L. Rose Bengal Labeled Bovine Serum Albumin for Protein Transport Imaging in Subcutaneous Tissues Using Computed Tomography and Fluorescence Microscopy. Bioconjug Chem 2024; 35:1044-1052. [PMID: 38875443 DOI: 10.1021/acs.bioconjchem.4c00240] [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: 06/16/2024]
Abstract
Subcutaneous (SC) injection of protein-based therapeutics is a convenient and clinically established drug delivery method. However, progress is needed to increase the bioavailability. Transport of low molecular weight (Mw) biotherapeutics such as insulin and small molecule contrast agents such as lipiodol has been studied using X-ray computed tomography (CT). This analysis, however, does not translate to the investigation of higher Mw therapeutics, such as monoclonal antibodies (mAbs), due to differences in molecular and formulation properties. In this study, an iodinated fluorescein analog rose bengal (RB) was used as a radiopaque and fluorescent label to track the distribution of bovine serum albumin (BSA) compared against unconjugated RB and sodium iodide (NaI) via CT and confocal microscopy following injection into ex vivo porcine SC tissue. Importantly, the high concentration BSA-RB exhibited viscosities more like that of viscous biologics than the small molecule contrast agents, suggesting that the labeled protein may serve as a more suitable formulation for the investigation of injection plumes. Three-dimensional (3D) renderings of the injection plumes showed that the BSA-RB distribution was markedly different from unconjugated RB and NaI, indicating the need for direct visualization of large protein therapeutics using conjugated tags rather than using small molecule tracers. Whereas this proof-of-concept study shows the novel use of RB as a label for tracking BSA distribution, our experimental approach may be applied to high Mw biologics, including mAbs. These studies could provide crucial information about diffusion in SC tissue and the influence of injection parameters on distribution, transport, and downstream bioavailability.
Collapse
Affiliation(s)
- Mazin H Hakim
- Weldon School of Biomedical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Melissa C Brindise
- School of Mechanical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Adib Ahmadzadegan
- School of Mechanical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Kevin P Buno
- Weldon School of Biomedical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Antonio C F Dos Santos
- Laboratory of Renewable Resources Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
- Department of Agricultural and Biological Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Kevin R Cragg
- Weldon School of Biomedical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Zhongwang Dou
- School of Mechanical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Michael R Ladisch
- Weldon School of Biomedical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
- Laboratory of Renewable Resources Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
- Department of Agricultural and Biological Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Pavlos P Vlachos
- Weldon School of Biomedical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
- School of Mechanical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
- Purdue Center for Cancer Research, Purdue University, 610 Purdue Mall, West Lafayette, Indiana 47907, United States
| |
Collapse
|
18
|
Tang Y, Liu B, Zhang Y, Liu Y, Huang Y, Fan W. Interactions between nanoparticles and lymphatic systems: Mechanisms and applications in drug delivery. Adv Drug Deliv Rev 2024; 209:115304. [PMID: 38599495 DOI: 10.1016/j.addr.2024.115304] [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: 12/12/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
The lymphatic system has garnered significant attention in drug delivery research due to the advantages it offers, such as enhancing systemic exposure and enabling lymph node targeting for nanomedicines via the lymphatic delivery route. The journey of drug carriers involves transport from the administration site to the lymphatic vessels, traversing the lymph before entering the bloodstream or targeting specific lymph nodes. However, the anatomical and physiological barriers of the lymphatic system play a pivotal role in influencing the behavior and efficiency of carriers. To expedite research and subsequent clinical translation, this review begins by introducing the composition and classification of the lymphatic system. Subsequently, we explore the routes and mechanisms through which nanoparticles enter lymphatic vessels and lymph nodes. The review further delves into the interactions between nanomedicine and body fluids at the administration site or within lymphatic vessels. Finally, we provide a comprehensive overview of recent advancements in lymphatic delivery systems, addressing the challenges and opportunities inherent in current systems for delivering macromolecules and vaccines.
Collapse
Affiliation(s)
- Yisi Tang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| | - Wufa Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
19
|
Shangguan W, Li X, Wang Y, Huang Z, Dong Y, Feng M, Feng J. Design and Biological Evaluation of the Long-Acting C5-Inhibited Ornithodoros moubata Complement Inhibitor (OmCI) Modified with Fatty Acid. Bioconjug Chem 2024; 35:653-664. [PMID: 38593046 DOI: 10.1021/acs.bioconjchem.4c00126] [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: 04/11/2024]
Abstract
Disorder of complement response is a significant pathogenic factor causing some autoimmune and inflammation diseases. The Ornithodoros moubata Complement Inhibitor (OmCI), a small 17 kDa natural protein, was initially extracted from soft tick salivary glands. The protein was found binding to complement C5 specifically, inhibiting the activation of the complement pathway, which is a successful therapeutic basis of complement-mediated diseases. However, a short half-life due to rapid renal clearance is a common limitation of small proteins for clinical application. In this study, we extended the half-life of OmCI by modifying it with fatty acid, which was a method used to improve the pharmacokinetics of native peptides and proteins. Five OmCI mutants were initially designed, and single-site cysteine mutation was introduced to each of them. After purification, four OmCI mutants were obtained that showed similar in vitro biological activities. Three mutants of them were subsequently coupled with different fatty acids by nucleophilic substitution. In total, 15 modified derivatives were screened and tested for anticomplement activity in vitro. The results showed that coupling with fatty acid would not significantly affect their complement-inhibitory activity (CH50 and AH50). OmCIT90C-CM02 and OmCIT90C-CM05 were validated as the applicable OmCI bioconjugates for further pharmacokinetic assessments, and both showed improved plasma half-life in mice compared with unmodified OmCI (15.86, 17.96 vs 2.57 h). In summary, our data demonstrated that OmCI conjugated with fatty acid could be developed as the potential long-acting C5 complement inhibitor in the clinic.
Collapse
Affiliation(s)
- Wenwen Shangguan
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Xiaowan Li
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Yandan Wang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 310014 Hangzhou, China
| | - Zongqing Huang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Yuanzhen Dong
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Meiqing Feng
- School of Pharmacy, Fudan University, 201203 Shanghai, China
| | - Jun Feng
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| |
Collapse
|
20
|
Rodríguez-Moranta F, Argüelles-Arias F, Hinojosa Del Val J, Iborra Colomino M, Martín-Arranz MD, Menchén Viso L, Muñoz Núñez F, Ricart Gómez E, Sánchez-Hernández JG, Valdés-Delgado T, Guardiola Capón J, Barreiro-de Acosta M, Mañosa Ciria M, Zabana Abdo Y, Gutiérrez Casbas A. Therapeutic drug monitoring in inflammatory bowel diseases. Position statement of the Spanish Working Group on Crohn's Disease and Ulcerative Colitis. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:522-552. [PMID: 38311005 DOI: 10.1016/j.gastrohep.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/31/2023] [Accepted: 01/18/2024] [Indexed: 02/06/2024]
Abstract
The treatment of inflammatory bowel disease has undergone a significant transformation following the introduction of biologic drugs. Thanks to these drugs, treatment goals have evolved from clinical response and remission to more ambitious objectives, such as endoscopic or radiologic remission. However, even though biologics are highly effective, a significant percentage of patients will not achieve an initial response or may lose it over time. We know that there is a direct relationship between the trough concentrations of the biologic and its therapeutic efficacy, with more demanding therapeutic goals requiring higher drug levels, and inadequate exposure being common. Therapeutic drug monitoring of biologic medications, along with pharmacokinetic models, provides us with the possibility of offering a personalized approach to treatment for patients with IBD. Over the past few years, relevant information has accumulated regarding its utility during or after induction, as well as in the maintenance of biologic treatment, in reactive or proactive strategies, and prior to withdrawal or treatment de-escalation. The aim of this document is to establish recommendations regarding the utility of therapeutic drug monitoring of biologics in patients with inflammatory bowel disease, in different clinical practice scenarios, and to identify areas where its utility is evident, promising, or controversial.
Collapse
Affiliation(s)
- Francisco Rodríguez-Moranta
- Servicio de Aparato Digestivo, Hospital Universitario de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, España.
| | - Federico Argüelles-Arias
- Servicio de Aparato Digestivo, Hospital Universitario Virgen Macarena, Sevilla, España; Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | | | - Marisa Iborra Colomino
- Servicio de Aparato Digestivo, Hospital Universitario y Politécnico de La Fe, Valencia, España
| | - M Dolores Martín-Arranz
- Servicio de Aparato Digestivo, Hospital Universitario La Paz, Facultad de Medicina de la UAM, Fundación para la investigación del Hospital Universitario la Paz (IDIPAZ), Madrid, España
| | - Luis Menchén Viso
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-IiSGM, Madrid, España; Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| | - Fernando Muñoz Núñez
- Servicio de Aparato Digestivo, Hospital Universitario de Salamanca, Salamanca, España
| | - Elena Ricart Gómez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), H. Clínic Barcelona, Barcelona, IDIBAPS, Barcelona, España
| | | | - Teresa Valdés-Delgado
- Servicio de Aparato Digestivo, Hospital Universitario Virgen Macarena, Sevilla, España
| | - Jordi Guardiola Capón
- Servicio de Gastroenterología, Hospital Universitario de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, España
| | - Manuel Barreiro-de Acosta
- Servicio de Gastroenterología, Hospital Clínico Universitario de Santiago, A Coruña, España; Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), A Coruña, España
| | - Míriam Mañosa Ciria
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, España; Unidad de Enfermedad Inflamatoria Intestinal, Servicio de Gastroenterología, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, España
| | - Yamile Zabana Abdo
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, España; Unidad de Enfermedad Inflamatoria Intestinal, Servicio de Gastroenterología, Hospital Mútua de Terrassa (HMT), Terrassa, Barcelona, España
| | - Ana Gutiérrez Casbas
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, España; Hospital General Universitario de Alicante, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, España
| |
Collapse
|
21
|
Asar M, Newton-Northup J, Soendergaard M. Improving Pharmacokinetics of Peptides Using Phage Display. Viruses 2024; 16:570. [PMID: 38675913 PMCID: PMC11055145 DOI: 10.3390/v16040570] [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: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Phage display is a versatile method often used in the discovery of peptides that targets disease-related biomarkers. A major advantage of this technology is the ease and cost efficiency of affinity selection, also known as biopanning, to identify novel peptides. While it is relatively straightforward to identify peptides with optimal binding affinity, the pharmacokinetics of the selected peptides often prove to be suboptimal. Therefore, careful consideration of the experimental conditions, including the choice of using in vitro, in situ, or in vivo affinity selections, is essential in generating peptides with high affinity and specificity that also demonstrate desirable pharmacokinetics. Specifically, in vivo biopanning, or the combination of in vitro, in situ, and in vivo affinity selections, has been proven to influence the biodistribution and clearance of peptides and peptide-conjugated nanoparticles. Additionally, the marked difference in properties between peptides and nanoparticles must be considered. While peptide biodistribution depends primarily on physiochemical properties and can be modified by amino acid modifications, the size and shape of nanoparticles also affect both absorption and distribution. Thus, optimization of the desired pharmacokinetic properties should be an important consideration in biopanning strategies to enable the selection of peptides and peptide-conjugated nanoparticles that effectively target biomarkers in vivo.
Collapse
Affiliation(s)
- Mallika Asar
- College of Osteopathic Medicine, Kansas City University, Kansas City, MO 64106, USA;
| | | | - Mette Soendergaard
- Cell Origins LLC, 1601 South Providence Road Columbia, Columbia, MO 65203, USA;
- Department of Chemistry, Western Illinois University, Macomb, IL 61455, USA
| |
Collapse
|
22
|
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
|
23
|
Walling DP, Shinde SN, Pogoda JM, Kharidia J, Laffont CM. An Open-Label Study to Assess Monthly Risperidone Injections (180 mg) Following Switch from Daily Oral Risperidone (6 mg) in Stable Schizophrenic Patients. Clin Drug Investig 2024; 44:251-260. [PMID: 38388986 PMCID: PMC10980608 DOI: 10.1007/s40261-024-01347-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND AND OBJECTIVE Long-acting injectable antipsychotics have shown benefits over oral medications with reduced hospitalization rates and improved health-related quality of life. RBP-7000 (PERSERIS®) is a monthly risperidone formulation (90 or 120 mg) for the treatment of schizophrenia administered by subcutaneous abdominal injection. The objective of this study was to assess a higher dose of 180 mg RBP-7000 and an alternate injection site. METHODS Following stabilization on 6 mg/day (3 mg twice daily) oral risperidone, clinically stable schizophrenic participants received 3 monthly doses of 180 mg RBP-7000 in the abdomen followed by a fourth monthly dose of 180 mg RBP-7000 in the upper arm (each dose administered as two 90-mg injections). The primary endpoint was the steady-state average plasma concentration (Cavg(ss)) of risperidone and total active moiety after oral and RBP-7000 administration. Secondary endpoints included measures of clinical efficacy (Positive and Negative Syndrome Scale, Clinical Global Impression Scale for Severity of Illness), safety, and local injection-site tolerability to assess the switch from oral risperidone and compare injection sites. RESULTS In all, 23 participants received at least one dose of RBP-7000, 16 received all four doses, and 15 completed the study. Monthly doses of 180 mg RBP-7000 provided similar Cavg(ss) of total active moiety compared with 6 mg/day oral risperidone. The pharmacokinetics of RBP-7000 were similar after injection in the abdomen versus upper arm. Clinical efficacy measures remained stable throughout the study. All RBP-7000 injections were well tolerated with no unexpected safety findings. CONCLUSIONS The results support the use of 180 mg RBP-7000 in schizophrenic patients stable on 6 mg/day oral risperidone and a second injection site in the upper arm. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03978832.
Collapse
Affiliation(s)
- David P Walling
- Collaborative Neuroscience Network, LLC, 12772 Valley View St. Ste 3, Garden Grove, CA, 92845, USA
| | - Sunita N Shinde
- Research and Development, Indivior Inc., 10710 Midlothian Turnpike, North Chesterfield, VA, 23235, USA
| | - Janice M Pogoda
- Research and Development, Indivior Inc., 10710 Midlothian Turnpike, North Chesterfield, VA, 23235, USA
| | - Jahnavi Kharidia
- Research and Development, Indivior Inc., 10710 Midlothian Turnpike, North Chesterfield, VA, 23235, USA
| | - Celine M Laffont
- Research and Development, Indivior Inc., 10710 Midlothian Turnpike, North Chesterfield, VA, 23235, USA.
| |
Collapse
|
24
|
Kim A, Oh MS, Lee GH, Song S, Byun MS, Choi D, Yu BY, Lee H. Understanding the pharmacokinetic journey of Fc-fusion protein, rhIL-7-hyFc using complementary approach of two analytical methods, accelerator mass spectrometry and ELISA. Antib Ther 2024; 7:105-113. [PMID: 38566969 PMCID: PMC10983079 DOI: 10.1093/abt/tbae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/21/2024] [Accepted: 01/28/2024] [Indexed: 04/04/2024] Open
Abstract
Antibody-based therapeutics (ABTs), including monoclonal/polyclonal antibodies and fragment crystallizable region (Fc)-fusion proteins, are increasingly used in disease treatment, driving the global market growth. Understanding the pharmacokinetic (PK) properties of ABTs is crucial for their clinical effectiveness. This study investigated the PK profile and tissue distribution of efineptakin alfa, a long-acting recombinant human interleukin-7 (rhIL-7-hyFc), using enzyme-linked immunosorbent assay (ELISA) and accelerator mass spectrometry (AMS). Totally, four rats were injected intramuscularly with 1 mg/kg of rhIL-7-hyFc containing 14C-rhIL-7-hyFc, which was prepared via reductive methylation. Serum total radioactivity (TRA) and serum rhIL-7-hyFc concentrations were quantified using AMS and ELISA, respectively. The TRA concentrations in organs were determined by AMS. Serum TRA peaked at 10 hours with a terminal half-life of 40 hours. The rhIL-7-hyFc exhibited a mean peak concentration at around 17 hours and a rapid elimination with a half-life of 12.3 hours. Peak concentration and area under the curve of TRA were higher than those of rhIL-7-hyFc. Tissue distribution analysis showed an elevated TRA concentrations in lymph nodes, kidneys, and spleen, indicating rhIL-7-hyFc's affinity for these organs. The study also simulated the positions of 14C labeling in rhIL-7-hyFc, identifying specific residues in the fragment of rhIL-7 portion, and provided the explanation of distinct analytes targeted by each method. Combining ELISA and AMS provided advantages by offering sensitivity and specificity for quantification as well as enabling the identification of analyte forms. The integrated use of ELISA and AMS offers valuable insights for the development and optimization of ABT.
Collapse
Affiliation(s)
- Anhye Kim
- Department of Clinical Pharmacology and Therapeutics, CHA Bundang Medical Center, CHA University, Seongnam 13496, Republic of Korea
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam 13488, Republic of Korea
- Institute for Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam 13488, Republic of Korea
| | - Min-Seok Oh
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Gwan-Ho Lee
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seongeun Song
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Mi-sun Byun
- Clinical Development Division, Genexine, Inc., Seoul 07789, Republic of Korea
| | - Donghoon Choi
- Research Institute, NeoImmuneTech, co. Ltd., Pohang 37666, Republic of Korea
| | - Byung-Yong Yu
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Howard Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Advanced Institute of Convergence Technology, Suwon 16229, Republic of Korea
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Mina-Osorio P, Tran MH, Habib AA. Therapeutic Plasma Exchange Versus FcRn Inhibition in Autoimmune Disease. Transfus Med Rev 2024; 38:150767. [PMID: 37867088 DOI: 10.1016/j.tmrv.2023.150767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 10/24/2023]
Abstract
Therapeutic plasma exchange (TPE or PLEX) is used in a broad range of autoimmune diseases, with the goal of removing autoantibodies from the circulation. A newer approach for the selective removal of immunoglobulin G (IgG) antibodies is the use of therapeutic molecules targeting the neonatal Fc receptor (FcRn). FcRn regulates IgG recycling, and its inhibition results in a marked decrease in circulating autoantibodies of the IgG subtype. The difference between FcRn inhibition and PLEX is often questioned. With anti-FcRn monoclonal antibodies (mAbs) and fragments only recently entering this space, limited data are available regarding long-term efficacy and safety. However, the biology of FcRn is well understood, and mounting evidence regarding the efficacy, safety, and potential differences among compounds in development is available, allowing us to compare against nonselective plasma protein depletion methods such as PLEX. FcRn inhibitors may have distinct advantages and disadvantages over PLEX in certain scenarios. Use of PLEX is preferred over FcRn inhibition where removal of antibodies other than IgG or when concomitant repletion of missing plasma proteins is needed for therapeutic benefit. Also, FcRn targeting has not yet been studied for use in acute flares or crisis states of IgG-mediated diseases. Compared with PLEX, FcRn inhibition is associated with less invasive access requirements, more specific removal of IgG versus other immunoglobulins without a broad impact on circulating proteins, and any impacts on other therapeutic drug levels are restricted to other mAbs. In addition, the degree of IgG reduction is similar with FcRn inhibitors compared with that afforded by PLEX. Here we describe the scientific literature regarding the use of PLEX and FcRn inhibitors in autoimmune diseases and provide an expert discussion around the potential benefits of these options in varying clinical conditions and scenarios.
Collapse
Affiliation(s)
| | - Minh-Ha Tran
- Department of Pathology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ali A Habib
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
28
|
Kutahya ZO, Akyol BA, Deliklitas K, Gokbulut C. Plasma disposition and faecal excretion of eprinomectin following subcutaneous administration in Saanen and Alpine goats. Res Vet Sci 2024; 166:105091. [PMID: 37984218 DOI: 10.1016/j.rvsc.2023.105091] [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: 09/04/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/22/2023]
Abstract
Eprinomectin is extensively used in veterinary medicine, particularly in the treatment of internal and external parasites in livestock, including goats. The pharmacokinetic behavior of eprinomectin in plasma and faeces was studied after a single subcutaneous administration in two different goat breeds at a dose of 0.2 mg/kg body weight. The study was conducted on one-year-old female Saanen (n = 8) and Alpine (n = 8) goats in a parallel design. There were no significant differences between Saanen and Alpine goats on the peak plasma concentration (Cmax, 28.59 ± 7.46 ng/mL vs. 37.69 ± 14.89 ng/mL), area under the curve (AUC0-∞, 93.08 ± 11.66 ng.d/mL vs. 116.98 ± 48.36 ng.d/mL), area under the first moment curve (AUMC0-∞, 311.05 ± 67.23 ng.d2/mL vs. 348.25 ± 202.64 ng.d2/mL) and mean residence time (MRT, 3.24 ± 0.77 d vs. 2.74 ± 0.64 d) values. The plasma terminal half-life and the time to reach peak plasma concentration were significantly higher in Saanen goats (T1/2λz, 2.18 ± 0.43 d; Tmax, 1.21 ± 0.25 d) than in Alpine goats (T1/2λz, 1.66 ± 0.41 d; Tmax, 0.79 ± 0.25 d). The results revealed that the plasma concentration of eprinomectin did not differ depending on the breed in Saanen and Alpine goats. However, it was determined that the eprinomectin clearance from the body may vary depending on the breed in goats. The faecal eprinomectin concentration of Saanen and Alpine goats was 90 and 80 times higher than the plasma eprinomectin concentration, respectively. Although high faecal excretion of eprinomectin confers a high efficacy advantage against parasites in the gastrointestinal tract, it may pose an ecotoxicological risk to manure fauna and aquatic organisms with high susceptibility to this compound.
Collapse
Affiliation(s)
- Zeynep Ozdemir Kutahya
- Cukurova University, Faculty of Ceyhan Veterinary Medicine, Department of Veterinary Pharmacology and Toxicology, Adana, Türkiye
| | - Busra Aslan Akyol
- Balikesir University, Institute of Health Sciences, Department of Veterinary Pharmacology and Toxicology, CoHE 100/2000 Scholarship Holder, Balikesir, Türkiye
| | - Kubra Deliklitas
- Balikesir University, Institute of Health Sciences, Department of Veterinary Pharmacology and Toxicology, CoHE 100/2000 Scholarship Holder, Balikesir, Türkiye
| | - Cengiz Gokbulut
- Balikesir University, Faculty of Medicine, Department of Medical Pharmacology, Balikesir, Türkiye; Institute of Health Sciences, Department of Veterinary Pharmacology and Toxicology, Balikesir, Türkiye.
| |
Collapse
|
29
|
Bei R, Thomas J, Kapur S, Woldeyes M, Rauk A, Robarge J, Feng J, Abbou Oucherif K. Predicting the clinical subcutaneous absorption rate constant of monoclonal antibodies using only the primary sequence: a machine learning approach. MAbs 2024; 16:2352887. [PMID: 38745390 PMCID: PMC11110684 DOI: 10.1080/19420862.2024.2352887] [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: 03/03/2024] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
Subcutaneous injections are an increasingly prevalent route of administration for delivering biological therapies including monoclonal antibodies (mAbs). Compared with intravenous delivery, subcutaneous injections reduce administration costs, shorten the administration time, and are strongly preferred from a patient experience point of view. An understanding of the absorption process of a mAb from the injection site to the systemic circulation is critical to the process of subcutaneous mAb formulation development. In this study, we built a model to predict the absorption rate constant (ka), which denotes how fast a mAb is absorbed from the site of administration. Once trained, our model (enabled by the XGBoost algorithm in machine learning) can predict the ka of a mAb following a subcutaneous injection using in silico molecular properties alone (generated from the primary sequence). Our model does not need clinically observed plasma concentration-time data; this is a novel capability not previously achieved in predictive pharmacokinetic models. The model also showed improved performance when benchmarked against a recently reported mechanistic model that relied on clinical data to predict subcutaneous absorption of mAbs. We further interpreted the model to understand which molecular properties affect the absorption rate and showed that our findings are consistent with previous studies evaluating subcutaneous absorption through direct experimentation. Taken altogether, this study reports the development, validation, benchmarking, and interpretation of a model that can predict the clinical ka of a mAb using its primary sequence as the only input.
Collapse
Affiliation(s)
- Ronghua Bei
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Justin Thomas
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Shiven Kapur
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mahlet Woldeyes
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Adam Rauk
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jason Robarge
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jiangyan Feng
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Kaoutar Abbou Oucherif
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
30
|
Gou S, Lapteva M, Brusini R, Flegeau K, Bourdon F, Kaya G, Faivre J, Kalia YN. Development of an ex vivo porcine skin model for the preclinical evaluation of subcutaneously injected biomacromolecules. Int J Pharm 2023; 648:123562. [PMID: 37907142 DOI: 10.1016/j.ijpharm.2023.123562] [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: 09/18/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/02/2023]
Abstract
Subcutaneous administration is used to deliver systemically-acting biotherapeutics, e.g. antibodies, and locally-acting biomacromolecules, e.g. hyaluronic acid. However, few preclinical models are available to evaluate post-injection behaviour in the tissue microenvironment. In vivo animal studies are costly, time-consuming, and raise obvious ethical concerns. In vitro models are cost-efficient, high-throughput solutions, but cannot simulate complex skin structure and biological function. An ex vivo model (containing hypodermis) with an extended culture period that enabled longitudinal studies would be of great interest for both the pharmaceutical and cosmeceutical industries. We describe the development of one such ex vivo model, using viable full-thickness porcine skin. Structural integrity was evaluated using a histological scoring system: spongiosis and epidermal detachment were identified as discriminating parameters. Ki67 and Claudin-1 expression reported on epidermal cell proliferation and barrier function, respectively and their expression decreased as a function of incubation time. After optimization, the system was used to investigate the fate/impact of subcutaneously administered hyaluronic acid (HA) formulations. The results showed that HA was localized at the injection site and adjacent adipocytes were well preserved during 5 days' incubation and confirmed that the full-thickness ex vivo porcine skin model could provide a platform for preclinical evaluation of subcutaneously injected biomacromolecules.
Collapse
Affiliation(s)
- Si Gou
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Maria Lapteva
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | | | | | | | - Gürkan Kaya
- Department of Dermatology, University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Jimmy Faivre
- Teoxane SA, Rue de Lyon 105, 1203 Geneva, Switzerland
| | - Yogeshvar N Kalia
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
31
|
Ng F, Nicoulin V, Peloso C, Curia S, Richard J, Lopez-Noriega A. In Vitro and In Vivo Hydrolytic Degradation Behaviors of a Drug-Delivery System Based on the Blend of PEG and PLA Copolymers. ACS APPLIED MATERIALS & INTERFACES 2023; 15:55495-55509. [PMID: 38011651 DOI: 10.1021/acsami.2c13141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
This paper presents the in vitro and in vivo degradation of BEPO, a marketed in situ forming depot technology used for the formulation of long-acting injectables. BEPO is composed of a solution of a blend of poly(ethylene glycol)-block-poly(lactic acid) (PEG-PLA) triblock and diblock in an organic solvent, where a therapeutic agent may be dissolved or suspended. Upon contact with an aqueous environment, the solvent diffuses and the polymers precipitate, entrapping the drug and forming a reservoir. Two representative BEPO compositions were subjected to a 3-month degradation study in vitro by immersion in phosphate-buffered saline at 37 °C and in vivo after subcutaneous injection in minipig. The material erosion rate, as a surrogate of the bioresorption, determined via the depot weight loss, changed substantially, depending on the composition and content of polymers within the test item. The swelling properties and internal morphology of depots were shown to be highly dependent on the solvent exchange rate during the precipitation step. Thermal analyses displayed an increase of the depot glass transition temperature over the degradation process, with no crystallinity observed at any stage. The chemical composition of degraded depots was determined by 1H NMR and gel permeation chromatography and demonstrated an enrichment in homopolymers, i.e., free PLA and (m)PEG, to the detriment of (m)PEG-PLA copolymers in both formulations. It was observed that the relative ratio of the degradants within the depot is driven by the initial polymer composition. Interestingly, in vitro and in vivo results showed very good qualitative consistency. Taken together, the outcomes from this study demonstrate that the different hydrolytic degradation behaviors of the BEPO compositions can be tuned by adjusting the polymer composition of the formulation.
Collapse
Affiliation(s)
- Feifei Ng
- MedinCell S.A., 3 Rue des Frères Lumière, 34830 Jacou, France
| | - Victor Nicoulin
- MedinCell S.A., 3 Rue des Frères Lumière, 34830 Jacou, France
| | | | - Silvio Curia
- MedinCell S.A., 3 Rue des Frères Lumière, 34830 Jacou, France
| | - Joël Richard
- MedinCell S.A., 3 Rue des Frères Lumière, 34830 Jacou, France
| | | |
Collapse
|
32
|
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
|
33
|
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: 0] [Impact Index Per Article: 0] [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
|
34
|
Jacquemier P, Retory Y, Virbel-Fleischman C, Schmidt A, Ostertag A, Cohen-Solal M, Alzaid F, Potier L, Julla JB, Gautier JF, Venteclef N, Riveline JP. New ex vivo method to objectively assess insulin spatial subcutaneous dispersion through time during pump basal-rate based administration. Sci Rep 2023; 13:20052. [PMID: 37973963 PMCID: PMC10654403 DOI: 10.1038/s41598-023-46993-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
Glycemic variability remains frequent in patients with type 1 diabetes treated with insulin pumps. Heterogeneous spreads of insulin infused by pump in the subcutaneous (SC) tissue are suspected but were barely studied. We propose a new real-time ex-vivo method built by combining high-precision imaging with simultaneous pressure measurements, to obtain a real-time follow-up of insulin subcutaneous propagation. Human skin explants from post-bariatric surgery are imaged in a micro-computed tomography scanner, with optimised parameters to reach one 3D image every 5 min during 3 h of 1UI/h infusion. Pressure inside the tubing is recorded. A new index of dispersion (IoD) is introduced and computed upon the segmented 3D insulin depot per time-step. Infusions were hypodermal in 58.3% among 24 assays, others being intradermal or extradermal. Several minor bubbles and one occlusion were observed. IoD increases with time for all injections. Inter-assay variability is the smallest for hypodermal infusions. Pressure elevations were observed, synchronised with air bubbles arrivals in the tissue. Results encourage the use of this method to compare infusion parameters such as pump model, basal rate, catheter characteristics, infusion site characteristics or patient phenotype.
Collapse
Affiliation(s)
- Pauline Jacquemier
- Institut Necker Enfants Malades (INEM), IMMEDIAB Laboratory, Université de Paris Cité, INSERM U1151, Paris, France
- Centre Explor, ALHIST - Air Liquide Healthcare, Bagneux, France
| | - Yann Retory
- LVL Médical Groupe, Lyon, France
- CIAMS, Univ. Paris-Sud, Université Paris-Saclay, 91405, Orsay Cedex, France
- CIAMS, Université d'Orléans, 45067, Orléans, France
| | | | | | - Agnes Ostertag
- Université Paris Cité, Inserm U1132 BIOSCAR, 75010, Paris, France
| | - Martine Cohen-Solal
- Université Paris Cité, Inserm U1132 BIOSCAR, 75010, Paris, France
- Service de Rhumatologie, Lariboisiere Hospital, AP-HP, 75010, Paris, France
| | - Fawaz Alzaid
- Institut Necker Enfants Malades (INEM), IMMEDIAB Laboratory, Université de Paris Cité, INSERM U1151, Paris, France
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Louis Potier
- Institut Necker Enfants Malades (INEM), IMMEDIAB Laboratory, Université de Paris Cité, INSERM U1151, Paris, France
- Université Paris Cité, UFR de Médecine, Paris, France
- Department of Diabetology, Endocrinology and Nutrition, Bichat Hospital, APHP, Paris, France
| | - Jean-Baptiste Julla
- Institut Necker Enfants Malades (INEM), IMMEDIAB Laboratory, Université de Paris Cité, INSERM U1151, Paris, France
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie Et de Santé Publique, 75013, Paris, France
- Service of Diabetology, Endocrinology and Nutrition, Federation de Diabetologie, Lariboisiere Hospital, 2 Rue Ambroise Paré, 75010, Paris, AP-HP, France
| | - Jean-François Gautier
- Institut Necker Enfants Malades (INEM), IMMEDIAB Laboratory, Université de Paris Cité, INSERM U1151, Paris, France
- Université Paris Cité, UFR de Médecine, Paris, France
- Service of Diabetology, Endocrinology and Nutrition, Federation de Diabetologie, Lariboisiere Hospital, 2 Rue Ambroise Paré, 75010, Paris, AP-HP, France
| | - Nicolas Venteclef
- Institut Necker Enfants Malades (INEM), IMMEDIAB Laboratory, Université de Paris Cité, INSERM U1151, Paris, France
| | - Jean-Pierre Riveline
- Institut Necker Enfants Malades (INEM), IMMEDIAB Laboratory, Université de Paris Cité, INSERM U1151, Paris, France.
- Université Paris Cité, UFR de Médecine, Paris, France.
- Service of Diabetology, Endocrinology and Nutrition, Federation de Diabetologie, Lariboisiere Hospital, 2 Rue Ambroise Paré, 75010, Paris, AP-HP, France.
| |
Collapse
|
35
|
Chua L, Friedrich S, Zhang XC. Mirikizumab Pharmacokinetics in Patients with Moderately to Severely Active Ulcerative Colitis: Results from Phase III LUCENT Studies. Clin Pharmacokinet 2023; 62:1479-1491. [PMID: 37610533 PMCID: PMC10520102 DOI: 10.1007/s40262-023-01281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND AND OBJECTIVE Mirikizumab is a humanized anti-interleukin-23-p19 monoclonal antibody being developed for ulcerative colitis and Crohn's disease. This analysis characterized mirikizumab pharmacokinetics using phase II and III trial data from patients with moderately to severely active ulcerative colitis. METHODS Serum pharmacokinetic data in patients receiving mirikizumab 50-1000 mg intravenously every 4 weeks as induction treatment and mirikizumab 200 mg subcutaneously every 4 or 12 weeks as maintenance treatment across three trials (N = 1362) were analyzed using non-linear mixed-effects modeling. Covariate effects on mirikizumab exposure were evaluated using simulation-based estimations. RESULTS Mirikizumab pharmacokinetics was best described by a linear two-compartment model with first-order absorption. Clearance, volume of distribution for central and peripheral compartments, and half-life were estimated at approximately 0.022 L/h (linear), 3.11 L and 1.69 L, and 9.5 days, respectively. Statistically significant effects of body weight and serum albumin levels on clearance, body weight on central and peripheral volumes of distribution, and body mass index on bioavailability were observed but effects were small relative to random inter-individual variability (% coefficient of variation: 18-64%). The subcutaneous bioavailability of mirikizumab was 48%. CONCLUSIONS Mirikizumab displayed pharmacokinetic characteristics typical of a monoclonal antibody where clearance increased with body weight and decreased with the albumin level, and bioavailability decreased with body mass index. These effects were small relative to random variability, indicating that a dose adjustment for patient factors is not required. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov: NCT02589665 (28 October, 2015), NCT03518086 (8 May, 2018), NCT03524092 (14 May, 2018).
Collapse
Affiliation(s)
- Laiyi Chua
- Global PK/PD and Pharmacometrics, Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN, 46225, USA
| | - Stuart Friedrich
- Global PK/PD and Pharmacometrics, Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN, 46225, USA
| | - Xin Cindy Zhang
- Global PK/PD and Pharmacometrics, Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN, 46225, USA.
| |
Collapse
|
36
|
Li C, Liu H, Liao Y, Zhu Y, Tian J, Wang X, Hu Z, Zhan Y, Li X, Liang X, He J, Li Y, Shang D, Zheng Q, Wang T, Song H, Fang Y. Phase I, Randomized, Placebo-Controlled, Dose-Escalation Study of GB223, a Fully-Humanized Monoclonal Antibody to RANKL, in Healthy Chinese Adults. BioDrugs 2023; 37:721-735. [PMID: 37278972 DOI: 10.1007/s40259-023-00604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND GB223 is a novel, fully-humanized monoclonal antibody against the receptor activator of nuclear factor-kappa B ligand (RANKL). In this phase I study, the safety, tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity of GB223 were investigated. PATIENTS AND METHODS This was a randomized, double-blinded, placebo-controlled, single-dose escalation study conducted in 44 healthy Chinese adults. Participants were randomly assigned to receive a single subcutaneous injection dose of 7, 21, 63, 119, or 140 mg of GB223 (n = 34) or placebo (n = 10) and were followed up for 140-252 days. RESULTS The results of noncompartmental analysis showed that GB223 was slowly absorbed after dosing, with a time to reach maximum concentration (Tmax) ranging from 5 to 11 days. Serum GB223 concentrations decreased slowly, with a long half-life ranging from 7.91 to 19.60 days. A two-compartment Michaelis-Menten model was found to best describe the pharmacokinetics of GB223, and the absorption rate of GB223 differed between males (0.0146 h-1) and females (0.0081 h-1). Serum C-terminal telopeptide of type I collagen decreased significantly postdose, and the inhibition lasted 42-168 days. No deaths or drug-related serious adverse events occurred. The most frequent adverse events were blood parathyroid hormone increased (94.1%), blood phosphorus decreased (67.6%) and blood calcium decreased (58.8%). In the GB223 group, 44.1% (15/34) of subjects were antidrug antibody positive after dosing. CONCLUSION In this study, we demonstrated for the first time that a single subcutaneous injection of GB223, from 7 to 140 mg, is safe and well tolerated in healthy Chinese subjects. GB223 has a nonlinear pharmacokinetic profile, and sex was a potential covariate that may affect the absorption rate of GB223. CLINICAL TRIAL REGISTRATION NCT04178044 and ChiCTR1800020338.
Collapse
Affiliation(s)
- Chen Li
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Phase I Clinical Research Center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haiyan Liu
- Phase I Clinical Research Center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yixiang Liao
- Phase I Clinical Research Center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu Zhu
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingyuan Tian
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuan Wang
- Phase I Clinical Research Center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiqin Hu
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaoxuan Zhan
- Phase I Clinical Research Center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xianbo Li
- Phase I Clinical Research Center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xintong Liang
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin He
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongmei Li
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dewei Shang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingshan Zheng
- Center for Drug Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tenghua Wang
- Department of Pharmacy, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Haifeng Song
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Yi Fang
- Clinical Trial Institution Research Ward, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
37
|
Jones G, Zeng L, Kim J. Application of Allometric Scaling to Nanochelator Pharmacokinetics. ACS OMEGA 2023; 8:27256-27263. [PMID: 37546686 PMCID: PMC10399172 DOI: 10.1021/acsomega.3c02570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/22/2023] [Indexed: 08/08/2023]
Abstract
Deferoxamine (DFO) is an effective FDA-approved iron chelator; however, its use is considerably limited by off-target toxicities and an extremely cumbersome dose regimen involving daily infusions. The recent development of a deferoxamine-based nanochelator (DFO-NP) with selective renal excretion has shown promise in ameliorating iron overload and associated physiological complications in rodent models with a substantially improved safety profile. While the dose- and administration route-dependent pharmacokinetics (PK) of DFO-NPs have been recently characterized, the optimized PK model was not validated, and the prior studies did not directly address the clinical translatability of DFO-NPs into humans. In the present work, these gaps were addressed by applying allometric scaling of DFO-NP PK in rats to predict those in mice and humans. First, this approach predicted serum concentration-time profiles of DFO-NPs, which were similar to those experimentally measured in mice, validating the nonlinear disposition and absorption models for DFO-NPs across the species. Subsequently, we explored the utility of allometric scaling by predicting the PK profile of DFO-NPs in humans under clinically relevant dosing schemes. These in silico efforts demonstrated that the novel nanochelator is expected to improve the PK of DFO when compared to standard infusion regimens of native DFO. Moreover, reasonable formulation strategies were identified and discussed for both early clinical development and more sophisticated formulation development.
Collapse
Affiliation(s)
- Gregory Jones
- Department
of Pharmaceutical Sciences, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Lingxue Zeng
- Department
of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| | - Jonghan Kim
- Department
of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| |
Collapse
|
38
|
Eke AC, Gebreyohannes RD, Fernandes MFS, Pillai VC. Physiologic Changes During Pregnancy and Impact on Small-Molecule Drugs, Biologic (Monoclonal Antibody) Disposition, and Response. J Clin Pharmacol 2023; 63 Suppl 1:S34-S50. [PMID: 37317492 PMCID: PMC10365893 DOI: 10.1002/jcph.2227] [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: 10/26/2022] [Accepted: 02/17/2023] [Indexed: 06/16/2023]
Abstract
Pregnancy is a unique physiological state that results in many changes in bodily function, including cellular, metabolic, and hormonal changes. These changes can have a significant impact on the way small-molecule drugs and monoclonal antibodies (biologics) function and are metabolized, including efficacy, safety, potency, and adverse effects. In this article, we review the various physiologic changes that occur during pregnancy and their effects on drug and biologic metabolism, including changes in the coagulation, gastrointestinal, renal, endocrine, hepatic, respiratory, and cardiovascular systems. Additionally, we discuss how these changes can affect the processes of drug and biologic absorption, distribution, metabolism, and elimination (pharmacokinetics), and how drugs and biologics interact with biological systems, including mechanisms of drug action and effect (pharmacodynamics) during pregnancy, as well as the potential for drug-induced toxicity and adverse effects in the mother and developing fetus. The article also examines the implications of these changes for the use of drugs and biologics during pregnancy, including consequences of suboptimal plasma drug concentrations, effect of pregnancy on the pharmacokinetics and pharmacodynamics of biologics, and the need for careful monitoring and individualized drug dosing. Overall, this article aims to provide a comprehensive understanding of the physiologic changes during pregnancy and their effects on drug and biologic metabolism to improve the safe and effective use of drugs.
Collapse
Affiliation(s)
- Ahizechukwu C Eke
- Division of Maternal Fetal Medicine, Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rahel D Gebreyohannes
- Department of Obstetrics and Gynecology, Addis Ababa University College of Medicine, Addis Ababa, Ethiopia
| | | | | |
Collapse
|
39
|
Rama B, Ribeiro AJ. Role of nanotechnology in the prolonged release of drugs by the subcutaneous route. Expert Opin Drug Deliv 2023; 20:559-577. [PMID: 37305971 DOI: 10.1080/17425247.2023.2214362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/11/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Subcutaneous physiology is distinct from other parenteral routes that benefit the administration of prolonged-release formulations. A prolonged-release effect is particularly convenient for treating chronic diseases because it is associated with complex and often prolonged posologies. Therefore, drug-delivery systems focused on nanotechnology are proposed as alternatives that can overcome the limitations of current therapeutic regimens and improve therapeutic efficacy. AREAS COVERED This review presents an updated systematization of nanosystems, focusing on their applications in highly prevalent chronic diseases. Subcutaneous-delivered nanosystem-based therapies comprehensively summarize nanosystems, drugs, and diseases and their advantages, limitations, and strategies to increase their translation into clinical applications. An outline of the potential contribution of quality-by-design (QbD) and artificial intelligence (AI) to the pharmaceutical development of nanosystems is presented. EXPERT OPINION Although recent academic research and development (R&D) advances in the subcutaneous delivery of nanosystems have exhibited promising results, pharmaceutical industries and regulatory agencies need to catch up. The lack of standardized methodologies for analyzing in vitro data from nanosystems for subcutaneous administration and subsequent in vivo correlation limits their access to clinical trials. There is an urgent need for regulatory agencies to develop methods that faithfully mimic subcutaneous administration and specific guidelines for evaluating nanosystems.
Collapse
Affiliation(s)
- B Rama
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
| | - A J Ribeiro
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
- Genetics of Cognitive Disfunction, i3S, IBMC, Porto, Portugal
| |
Collapse
|
40
|
Zhong X, Veilleux JC, Shi GH, Collins DS, Vlachos P, Ardekani AM. Hydrodynamic considerations for spring-driven autoinjector design. Int J Pharm 2023; 640:122975. [PMID: 37116602 DOI: 10.1016/j.ijpharm.2023.122975] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 04/30/2023]
Abstract
In recent years, significant progress has been made in the studies of the spring-driven autoinjector, leading to an improved understanding of this device and its interactions with tissue and therapeutic proteins. The development of simulation tools that have been validated against experiments has also enhanced the prediction of the performance of spring-driven autoinjectors. This paper aims to address critical hydrodynamic considerations that impact the design of spring-driven autoinjectors, with a specific emphasis on sloshing and cavitation. Additionally, we present a framework that integrates simulation tools to predict the performance of spring-driven autoinjectors and optimize their design. This work is valuable to the pharmaceutic industry, as it provides crucial insights into the development of spring-driven autoinjectors and therapeutic proteins. This work can also enhance the efficacy and safety of the delivery of therapeutic proteins, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Xiaoxu Zhong
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States
| | | | | | - David S Collins
- Eli Lilly and Company, Indianapolis, IN 46225, United States
| | - Pavlos Vlachos
- 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
|
41
|
Rau D, Eichau S, Borriello G, Cerqueira J, Wagner C. Assessment of the treating physicians' first-hand experience with handling and satisfaction of ofatumumab therapy: findings from the PERITIA survey conducted in Europe. BMC Neurol 2023; 23:147. [PMID: 37038104 PMCID: PMC10084644 DOI: 10.1186/s12883-023-03190-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/28/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Real-world evidence on experience and satisfaction of ofatumumab as a treatment option for relapsing multiple sclerosis (RMS) is limited. OBJECTIVE To present cumulative responses from a questionnaire related to first-hand experience of treating physicians on handling and convenience of ofatumumab therapy along with concerns related to COVID-19. METHODS PERITIA was a multicentre survey conducted to collect responses from the ASCLEPIOS I/II trial investigators from Europe via an online questionnaire. RESULTS Forty-six physicians (Germany, n = 14; Spain, n = 12; Portugal, n = 10; Italy, n = 10) completed the survey. Overall, 43% of the physicians considered the benefit-risk ratio of ofatumumab as very good. Over 93% were in favour of ofatumumab self-administration at home and the majority (83%) believed it to be completely true that self-administration of ofatumumab eases the burden for patients in terms of time. All investigators would like to potentially use anti-CD20 therapy as a long-term strategy. Even during the COVID-19 pandemic, physicians were in favour of a self-administration of MS therapy at home over other anti-CD20 therapy infusions. CONCLUSION European neurologists who were part of this survey considered the benefit-risk-ratio of ofatumumab as favourable and the monthly self-administered subcutaneous injections offering convenience for patients in the clinical practice.
Collapse
Affiliation(s)
| | - Sara Eichau
- Hospital Universitario Virgen de La Macareona of Sevilla, Seville, Spain
| | - Giovanna Borriello
- MS Center, Neurology Unit, Fatebenefratelli San Pietro Hospital, Rome, Italy
| | | | - Carola Wagner
- Novartis Pharma GmbH, Nuremberg, Germany and working on behalf of Novartis Pharma Vertriebs GmbH, Nuremberg, Germany
| |
Collapse
|
42
|
Zou P. Predicting Human Bioavailability of Subcutaneously Administered Fusion Proteins and Monoclonal Antibodies Using Human Intravenous Clearance or Antibody Isoelectric Point. AAPS J 2023; 25:31. [PMID: 36959523 DOI: 10.1208/s12248-023-00798-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/03/2023] [Indexed: 03/25/2023] Open
Abstract
There has been an increasing trend towards subcutaneous (SC) delivery of fusion proteins and monoclonal antibodies (mAbs) in recent years versus intravenous (IV) administration. The prediction of bioavailability is one of the major barriers in clinical translation of SC-administered therapeutic proteins due to a lack of reliable in vitro and preclinical in vivo predictive models. In this study, we explored the relationships between human SC bioavailability and physicochemical or pharmacokinetic properties of 19 Fc- or albumin-fusion proteins and 98 monoclonal antibodies. An inverse linear correlation was observed between human SC bioavailability and intravenous clearance (CL) or isoelectric point (pI). Multivariate regression models were developed using intravenous CL and pI of a training set (N = 59) as independent variables. The predictive models of mAbs were validated with an independent test set (N = 33). Two linear regression models resulted in 24 (73%) and 27 (82%) among 33 predictions within 0.8- to 1.2-fold deviations. Due to the small sample size of dataset, regression model validation was not conducted for fusion proteins. Overall, this study demonstrated that CL- and pI-based multivariate regression models could be used to predict human SC bioavailability of mAbs.
Collapse
Affiliation(s)
- Peng Zou
- Quantitative Clinical Pharmacology, Daiichi Sankyo, Inc., 211 Mt. Airy Road, Basking Ridge, New Jersey, 07920, USA.
| |
Collapse
|
43
|
Gao X, Sheng YH, Yu S, Li J, Rosa R, Girgis S, Guo T, Brunetti L, Kagan L. Mechanisms of Obesity-Induced Changes in Pharmacokinetics of IgG in Rats. Pharm Res 2023; 40:1223-1238. [PMID: 36949370 PMCID: PMC10033182 DOI: 10.1007/s11095-023-03496-y] [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: 02/02/2023] [Accepted: 03/01/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE To evaluate how obesity affects the pharmacokinetics of human IgG following subcutaneous (SC) and intravenous (IV) administration to rats and the homeostasis of endogenous rat IgG. METHODS Differences in body weight and size, body composition, and serum concentration of endogenous rat IgG in male Zucker obese (ZUC-FA/FA) and control (ZUC-LEAN) rats were measured from the age of 5 weeks up to 30 weeks. At the age of 23-24 weeks animals received a single IV or SC dose of human IgG (1 g/kg of total body weight), and serum pharmacokinetics was followed for 7 weeks. A mechanistic model linking obesity-related changes in pharmacokinetics with animal growth and changes in body composition was developed. RESULTS Significant differences were observed in both endogenous and exogenous IgG pharmacokinetics between obese and control groups. The AUC for human IgG was lower in obese groups (57.6% of control after IV and 48.1% after SC dosing), and clearance was 1.75-fold higher in obese animals. The mechanistic population model successfully captured the data and included several major components: endogenous rat IgG homeostasis with age-dependent synthesis rate; competition of human IgG and endogenous rat IgG for FcRn binding and its effect on endogenous rat IgG concentrations following injection of a high dose of human IgG; and the effect of body size and composition (changing over time and dependent on the obesity status) on pharmacokinetic parameters. CONCLUSIONS We identified important obesity-induced changes in the pharmacokinetics of IgG. Results can potentially facilitate optimization of the dosing of IgG-based therapeutics in the obese population.
Collapse
Affiliation(s)
- Xizhe Gao
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Yi-Hua Sheng
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Sijia Yu
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jiadong Li
- Comparative Medicine Resources, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Raymond Rosa
- Comparative Medicine Resources, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Simone Girgis
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Tiffany Guo
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Luigi Brunetti
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Pharmacy Practice and Administration, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Leonid Kagan
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
44
|
Imaging of large volume subcutaneous deposition using MRI: exploratory clinical study results. Drug Deliv Transl Res 2023:10.1007/s13346-023-01318-7. [PMID: 36913105 PMCID: PMC10382358 DOI: 10.1007/s13346-023-01318-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 03/14/2023]
Abstract
Subcutaneous (SC) delivery is a preferred route of administration for biotherapeutics but has predominantly been limited to volumes below 3 mL. With higher volume drug formulations emerging, understanding large volume SC (LVSC) depot localization, dispersion, and impact on the SC environment has become more critical. The aim of this exploratory clinical imaging study was to assess the feasibility of magnetic resonance imaging (MRI) to identify and characterize LVSC injections and their effect on SC tissue as a function of delivery site and volume. Healthy adult subjects received incremental injections of normal saline up to 5 mL total volume in the arm and up to 10 mL in the abdomen and thigh. MRI images were acquired after each incremental SC injection. Post-image analysis was performed to correct imaging artifacts, identify depot tissue location, create 3-dimensional (3D) SC depot rendering, and estimate in vivo bolus volumes and SC tissue distention. LVSC saline depots were readily achieved, imaged using MRI, and quantified via subsequent image reconstructions. Imaging artifacts occurred under some conditions, necessitating corrections applied during image analysis. 3D renderings were created for both the depot alone and in relation to the SC tissue boundaries. LVSC depots remained predominantly within the SC tissue and expanded with increasing injection volume. Depot geometry varied across injection sites and localized physiological structure changes were observed to accommodate LVSC injection volumes. MRI is an effective means to clinically visualize LVSC depots and SC architecture allowing assessment of deposition and dispersion of injected formulations.Trial Registration: Not applicable for this exploratory clinical imaging study.
Collapse
|
45
|
Ozer I, Slezak A, Sirohi P, Li X, Zakharov N, Yao Y, Everitt JI, Spasojevic I, Craig SL, Collier JH, Campbell JE, D'Alessio DA, Chilkoti A. An injectable PEG-like conjugate forms a subcutaneous depot and enables sustained delivery of a peptide drug. Biomaterials 2023; 294:121985. [PMID: 36630826 PMCID: PMC10918641 DOI: 10.1016/j.biomaterials.2022.121985] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023]
Abstract
Many biologics have a short plasma half-life, and their conjugation to polyethylene glycol (PEG) is commonly used to solve this problem. However, the improvement in the plasma half-life of PEGylated drugs' is at an asymptote because the development of branched PEG has only had a modest impact on pharmacokinetics and pharmacodynamics. Here, we developed an injectable PEG-like conjugate that forms a subcutaneous depot for the sustained delivery of biologics. The PEG-like conjugate consists of poly[oligo(ethylene glycol) methyl ether methacrylate] (POEGMA) conjugated to exendin, a peptide drug used in the clinic to treat type 2 diabetes. The depot-forming exendin-POEGMA conjugate showed greater efficacy than a PEG conjugate of exendin as well as Bydureon, a clinically approved sustained-release formulation of exendin. The injectable depot-forming exendin-POEGMA conjugate did not elicit an immune response against the polymer, so that it remained effective and safe for long-term management of type 2 diabetes upon chronic administration. In contrast, the PEG conjugate induced an anti-PEG immune response, leading to early clearance and loss of efficacy upon repeat dosing. The exendin-POEGMA depot also showed superior long-term efficacy compared to Bydureon. Collectively, these results suggest that an injectable POEGMA conjugate of biologic drugs that forms a drug depot under the skin, providing favorable pharmacokinetic properties and sustained efficacy while remaining non-immunogenic, offers significant advantages over other commonly used drug delivery technologies.
Collapse
Affiliation(s)
- Imran Ozer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Anna Slezak
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Parul Sirohi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Xinghai Li
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nikita Zakharov
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yunxin Yao
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Jeffrey I Everitt
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Ivan Spasojevic
- Duke School of Medicine, Department of Medicine-Oncology, Durham, NC, USA; Duke Cancer Institute, PK/PD Core Laboratory, Durham, NC, USA
| | | | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Division of Endocrinology, Duke University Medical Center, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - David A D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Division of Endocrinology, Duke University Medical Center, Durham, NC, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
46
|
Hakim MH, Jun BH, Ahmadzadegan A, Babiak PM, Xu Q, Buno KP, Liu JC, Ardekani AM, Vlachos PP, Solorio L. Investigation of macromolecular transport through tunable collagen hyaluronic acid matrices. Colloids Surf B Biointerfaces 2023; 222:113123. [PMID: 36640539 DOI: 10.1016/j.colsurfb.2023.113123] [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: 09/12/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
Therapeutic macromolecules possess properties such as size and electrostatic charge that will dictate their transport through subcutaneous (SC) tissue and ultimate bioavailability and efficacy. To improve therapeutic design, platforms that systematically measure the transport of macromolecules as a function of both drug and tissue properties are needed. We utilize a Transwell chamber with tunable collagen-hyaluronic acid (ColHA) hydrogels as an in vitro model to determine mass transport of macromolecules using non-invasive UV spectroscopy. Increasing hyaluronic acid (HA) concentration from 0 to 2 mg/mL within collagen gels decreases the mass transport of five macromolecules independent of size and charge and results in a maximum decrease in recovery of 23.3% in the case of bovine immunoglobulin G (IgG). However, in a pure 10 mg/mL HA solution, negatively-charged macromolecules bovine serum albumin (BSA), β-lactoglobulin (BLg), dextran (Dex), and IgG had drastically increased recovery by 20-40% compared to their performance in ColHA matrices. This result was different from the positively-charged macromolecule Lysozyme (Lys), which, despite its small size, showed reduced recovery by 3% in pure HA. These results demonstrate two distinct regimes of mass transport within our tissue model. In the presence of both collagen and HA, increasing HA concentrations decrease mass transport; however, in the absence of collagen, the high negative charge of HA sequesters and increases residence time of positively-charged macromolecules and decreases residence time of negatively-charged macromolecules. Through our approach, ColHA hydrogels serve as a platform for the systematic evaluation of therapeutic macromolecule transport as a function of molecular characteristics.
Collapse
Affiliation(s)
- Mazin H Hakim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Brian H Jun
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Adib Ahmadzadegan
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Qinghua Xu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Kevin P Buno
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Julie C Liu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Pavlos P Vlachos
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
47
|
Lymphatic uptake of biotherapeutics through a 3D hybrid discrete-continuum vessel network in the skin tissue. J Control Release 2023; 354:869-888. [PMID: 36634711 DOI: 10.1016/j.jconrel.2022.12.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 01/14/2023]
Abstract
Subcutaneous administration is a common approach for the delivery of biotherapeutics, which is achieved mainly through the absorption across lymphatic vessels. In this paper, the drug transport and lymphatic uptake through a three-dimensional hybrid discrete-continuum vessel network in the skin tissue are investigated through high-fidelity numerical simulations. We find that the local lymphatic uptake through the explicit vessels significantly affects macroscopic drug absorption. The diffusion of drug solute through the explicit vessel network affects the lymphatic uptake after the injection. This effect, however, cannot be captured using previously developed continuum models. The lymphatic uptake is dominated by the convection due to lymphatic drainage driven by the pressure difference, which is rarely studied in experiments and simulations. Furthermore, the effects of injection volume and depth on the lymphatic uptake are investigated in a multi-layered domain. We find that the injection volume significantly affects the rate of lymphatic uptake through the heterogeneous vessel network, while the injection depth has little influence, which is consistent with the experimental results. At last, the binding and metabolism of drug molecules are studied to bridge the simulations to the drug clearance experients. We provide a new approach to study the diffusion and convection of drug molecules into the lymphatic system through the hybrid vessel network.
Collapse
|
48
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
49
|
Sun J, Lin X, He Y, Zhang B, Zhou N, Huang JD. A bacterial outer membrane vesicle-based click vaccine elicits potent immune response against Staphylococcus aureus in mice. Front Immunol 2023; 14:1088501. [PMID: 36742310 PMCID: PMC9892643 DOI: 10.3389/fimmu.2023.1088501] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Staphylococcus aureus infection is a severe public health concern with the growing number of multidrug-resistant strains. S. aureus can circumvent the defense mechanisms of host immunity with the aid of multiple virulence factors. An efficacious multicomponent vaccine targeting diverse immune evasion strategies developed by S. aureus is thus crucial for its infection control. In this study, we exploited the SpyCatcher-SpyTag system to engineer bacterial outer membrane vesicles (OMVs) for the development of a multitargeting S. aureus click vaccine. We decorated OMVs with surface exposed SpyCatcher via a truncated OmpA(a.a 1-155)-SpyCatcher fusion. The engineered OMVs can flexibly bind with various SpyTag-fused S. aureus antigens to generate an OMV-based click vaccine. Compared with antigens mixed with alum adjuvant, the click vaccine simultaneously induced more potent antigen-specific humoral and Th1-based cellular immune response, which afforded protection against S. aureus Newman lethal challenge in a mouse model. Our study provided a flexible and versatile click vaccine strategy with the potential for fighting against emerging S. aureus clinical isolates.
Collapse
Affiliation(s)
- Jingjing Sun
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.,ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Xuansheng Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yige He
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Baozhong Zhang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Nan Zhou
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.,ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Jian-Dong Huang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China.,Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
50
|
Hu S, Datta-Mannan A, D'Argenio DZ. Monoclonal Antibody Pharmacokinetics in Cynomolgus Monkeys Following Subcutaneous Administration: Physiologically Based Model Predictions from Physiochemical Properties. AAPS J 2022; 25:5. [PMID: 36456779 DOI: 10.1208/s12248-022-00772-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/31/2022] [Indexed: 12/03/2022] Open
Abstract
An integrated physiologically based modeling framework is presented for predicting pharmacokinetics and bioavailability of subcutaneously administered monoclonal antibodies in cynomolgus monkeys, based on in silico structure-derived metrics characterizing antibody size, overall charge, local charge, and hydrophobicity. The model accounts for antibody-specific differences in pinocytosis, transcapillary transport, local lymphatic uptake, and pre-systemic degradation at the subcutaneous injection site and reliably predicts the pharmacokinetics of five different wild-type mAbs and their Fc variants following intravenous and subcutaneous administration. Significant associations were found between subcutaneous injection site degradation rate and the antibody's local positive charge of its complementarity-determining region (R = 0.56, p = 0.0012), antibody pinocytosis rate and its overall positive charge (R = 0.59, p = 0.00063), and antibody paracellular transport and its overall charge together with hydrophobicity (R = 0.63, p = 0.00096). Based on these results, population simulations were performed to predict the relationship between bioavailability and antibody local positive charge. In addition, model simulations were conducted to calculate the relative contribution of absorption pathways (lymphatic and blood), pre-systemic degradation pathways (interstitial and lysosomal), and the influence of injection site lymph flow on antibody bioavailability and pharmacokinetics. The proposed physiologically based modeling framework integrates fundamental mechanisms governing antibody subcutaneous absorption and disposition, with structured-based physiochemical properties, to predict antibody bioavailability and pharmacokinetics in vivo.
Collapse
Affiliation(s)
- Shihao Hu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, 90089, USA
| | - Amita Datta-Mannan
- Department of Exploratory Medicine and Pharmacology, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - David Z D'Argenio
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, 90089, USA.
| |
Collapse
|