1
|
Estupiñán HY, Baladi T, Roudi S, Munson MJ, Bost J, Gustafsson O, Velásquez-Ramírez D, Bhatt DK, Hagey D, Hekman D, Andersson S, Andaloussi SEL, Dahlén A. Design and screening of novel endosomal escape compounds that enhance functional delivery of oligonucleotides in vitro. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102522. [PMID: 40235852 PMCID: PMC11999280 DOI: 10.1016/j.omtn.2025.102522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/16/2025] [Indexed: 04/17/2025]
Abstract
Antisense oligonucleotides (ASOs), including splice-switching oligonucleotides (SSOs), are promising therapeutic approaches for targeting genetic defects. ASOs act in the nucleus and the cytosol to cleave mRNAs via the RNaseH1 mechanism (e.g., gapmers), while SSOs alter transcript splicing to restore or inhibit protein function. RNA interference (RNAi) is another approach to down-regulate gene expression via the RISC complex. However, a major challenge is the effective delivery of these nucleic acid-based therapeutics. Recent developments focus on enhancing cellular uptake and endosomal release, including the use of small-molecule endosomal escape enhancers (EEEs) such as chloroquine. Here, we disclose a next generation of EEEs, which efficiently enhance SSOs and gapmers in vitro activity. We identify proton sponge-mediated endosomal leakage as a mechanism of action and observe, by Gene Ontology analysis on bulk RNA sequencing, that EEE treatment increased gene expression of markers associated with vesicle organization. Additionally, using primary human hepatocytes, we demonstrate that EEEs enhance small interfering RNA (siRNA) activity. Unconjugated siRNA reached similar levels of mRNA knockdown to the observed GalNAc-conjugated siRNA. Substantial GalNAc conjugated siRNA enhancement was also observed when used together with EEE. Our results indicate that these EEEs constitute a promising strategy to enhance the activity of multimodal oligonucleotide therapeutics.
Collapse
Affiliation(s)
- H. Yesid Estupiñán
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Departamento de Ciencias Básicas, Universidad Industrial de Santander, Bucaramanga, Colombia
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Tom Baladi
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Samantha Roudi
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Michael J. Munson
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Jeremy Bost
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Oskar Gustafsson
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Daniel Velásquez-Ramírez
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Deepak Kumar Bhatt
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Hagey
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Dennis Hekman
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Shalini Andersson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Anders Dahlén
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
2
|
Tian Z, Luo H, Chu Y, Liu Y, Gao S, Song L, Yang Z, Liu D. Prediction of Interspecies Translation for Targeting Delivery Coefficients of GalNAc-siRNA Silencing Apolipoprotein C-III Using a Mechanistic Minimal Physiologically Based Pharmacokinetic/Pharmacodynamic Model. Clin Pharmacokinet 2025:10.1007/s40262-025-01513-4. [PMID: 40317426 DOI: 10.1007/s40262-025-01513-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND AND OBJECTIVE The emerging N-acetylgalactosamine-small interfering RNA (GalNAc-siRNA) conjugates lead the way for liver-targeting delivery to exert gene-silencing therapeutic effects. To facilitate the drug development of GalNAc-siRNA, further detailed understanding of the key modality-specific mechanisms underlying the temporal discordance between pharmacokinetics and pharmacodynamics and how these processes can be extrapolated from animals to humans is needed. METHODS A mechanistic minimal physiologically based pharmacokinetic/pharmacodynamic (mPBPK-PD) model for an investigational new apolipoprotein C-III (APOC3)-silencing GalNAc-siRNA (RBD5044) was developed using available pharmacokinetic/pharmacodynamic (PK/PD) data. The aim was to explore hepatic-targeting delivery processes, the PK/PD relationship, and interspecies translation. RESULTS First, multiple PK/PD datasets from mice were satisfactorily fitted using the mPBPK-PD model. Second, we translated the mice model to the monkey model, validated it, and then extrapolated from mice and monkeys to humans to simulate the PK/PD characteristics. We then mechanistically summarized and proposed the essential in vivo delivery processes of GalNAc-siRNA after subcutaneous administration (termed "ADUEB": Absorption [into system circulation], Disposition [distribution to liver target and elimination], Uptake [into hepatocytes], Escape [from endosome and lysosome compartments], and Binding [with argonaute2 to form RNA-induced silencing complex]). The targeting delivery coefficients of these processes achieved with the model using RBD5044 and the published data of another GalNAc-siRNA (fitusiran) quantitatively reflected the delivery efficiency and rate-limiting factors in targeted hepatocytes. CONCLUSION This study successfully constructed the mPBPK-PD model and conducted interspecies extrapolation for a GalNAc-siRNA targeting APOC3. Promising quantitative insights into a hepatic-targeted GalNAc-siRNA delivery system are provided to characterize the unique temporal disconnection of PK/PD properties and evaluate the key in vivo delivery processes. It will promote model-informed strategies and quantitative mechanistic understanding to support efficient drug development, evaluation, and clinical application of this modality in the future.
Collapse
Affiliation(s)
- Zhiteng Tian
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China
| | - Hui Luo
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Yantao Chu
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Yanhong Liu
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Shan Gao
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Ling Song
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.
- Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital Beijing, Beijing, 100191, China.
| | - Zhenzhen Yang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.
- Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital Beijing, Beijing, 100191, China.
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.
- Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital Beijing, Beijing, 100191, China.
| |
Collapse
|
3
|
Eladnani RP, Schaeper U, Diab R, Aretz J, Vrotniakaite-Bajerciene K, Çaku S, Yasmin R, Li B, Reina Caro MD, Dames S, Eisermann M, Löffler K, Martinez A, de Laat B, Brodard J, Casini A, Kremer Hovinga JA, Allam R, Fernández JA, Griffin JH, Laffan MA, Majumder R, Ahnström J, Angelillo-Scherrer A. Enhancing hemostasis potency in hemophilia with a small interfering ribonucleic acid targeting protein S. J Thromb Haemost 2025:S1538-7836(25)00201-6. [PMID: 40154791 DOI: 10.1016/j.jtha.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND One hemophilia treatment concept focuses on rebalancing coagulation and anticoagulation to restore normal blood clotting. Targeting the coagulation regulator, protein S (PS), in hemophilia shows promise to increase the generation of thrombin, a critical enzyme in the clotting process. OBJECTIVES This study aimed to: (1) assess whether inhibiting PS increases thrombin generation (TG) in plasma from individuals with hemophilia A (HA) and B (HB); and (2) develop a hepatocyte-targeted PS small interfering RNA (siRNA) therapy using N-acetylgalactosamine conjugation to restore hemostasis in hemophilia without increasing thromboembolic risks. METHODS We assessed TG in plasma from patients with HA and HB. To target the liver specifically, we developed a PS-siRNA conjugated with N-acetylgalactosamine. This approach ensures that PS levels remain adequate in other cells, thereby minimizing the risk of thrombosis. Additionally, we evaluated the therapeutic potential of PS-siRNA in preclinical models. RESULTS Inhibiting PS with a polyclonal antibody in plasma resulted in a 3- to 5-fold increase in TG in HA and a 4- to 9-fold increase in HB plasma, with a 70% reduction in plasma PS. In preclinical models, subcutaneous PS-siRNA therapy in HA mice and nonhuman primates successfully lowered PS levels and improved clot formation. It also prevented bleeding in both saphenous vein puncture and knee injury models in HA mice. Notably, it enhanced clotting without triggering widespread clot formation. CONCLUSION Reducing PS levels enhances TG in hemophilia models, and PS-siRNA therapy shows promise in improving hemostasis. This approach warrants further clinical investigation as a potential treatment for hemophilia.
Collapse
Affiliation(s)
- Raja Prince Eladnani
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Rim Diab
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Kristina Vrotniakaite-Bajerciene
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Sara Çaku
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Rafika Yasmin
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Bojun Li
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Maria Desiré Reina Caro
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | - Bas de Laat
- Synapse Research Institute, Maastricht, the Netherlands
| | - Justine Brodard
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alessandro Casini
- Division of Angiology and Hemostasis, University Hospitals of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Johanna A Kremer Hovinga
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ramanjaneyulu Allam
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - José A Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Mike A Laffan
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, London, UK
| | - Rinku Majumder
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Josefin Ahnström
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, London, UK
| | - Anne Angelillo-Scherrer
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
4
|
Choi GW, Kim JH, Kang DW, Cho HY. A journey into siRNA therapeutics development: A focus on Pharmacokinetics and Pharmacodynamics. Eur J Pharm Sci 2025; 205:106981. [PMID: 39643127 DOI: 10.1016/j.ejps.2024.106981] [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: 07/31/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
siRNA therapeutics are emerging novel modalities targeting highly specific mRNA via RNA interference mechanism. Its unique pharmacokinetics (PKs) and pharmacodynamics (PDs) are significant challenges for clinical use. Furthermore, naked siRNA is a highly soluble macromolecule with a negative charge, making plasma membrane penetration a significant hurdle. It is also vulnerable to nuclease degradation. Therefore, advanced formulation technologies, such as lipid nanoparticles and N-acetylgalactosamine conjugation, have been developed and are now used in clinical practice to enhance target organ delivery and stability. The innate complex biological mechanisms of siRNA, along with its formulation, are major determinants of the PK/PD characteristics of siRNA products. To systematically and quantitatively understand these characteristics, it is essential to develop and utilize quantitative PK/PD models for siRNA therapeutics. In this review, the effects of formulation on the PKs and PK/PD models of approved siRNA products were presented, highlighting the importance of selecting appropriate biomarkers and understanding formulation, PKs, and PDs for quantitative interpreting the relationship between plasma concentration, organ concentration, biomarkers, and efficacy.
Collapse
Affiliation(s)
- Go-Wun Choi
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Ju Hee Kim
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Dong Wook Kang
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Hea-Young Cho
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea.
| |
Collapse
|
5
|
Li Q, Geng T, Li H, Zheng S, Svedlund S, Gan L, Egnell AC, Gao S, Chen R, Hu P. Analysis of the pharmacokinetics and efficacy of RBD1016 - A GalNAc-siRNA targeting Hepatitis B Virus X gene using semi-mechanistic PK/PD model. Heliyon 2024; 10:e31924. [PMID: 38841435 PMCID: PMC11152740 DOI: 10.1016/j.heliyon.2024.e31924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
Small interference RNA (siRNA) is a class of short double-stranded RNA molecules that cause mRNA degradation through an RNA interference mechanism and is a promising therapeutic modality. RBD1016 is a siRNA drug in clinical development for the treatment of chronic Hepatitis B Virus (HBV) infection, which contains a conjugated with N-acetylglucosamine moiety that can facilitate its hepatic delivery. We aimed to construct a semi-mechanistic model of RBD1016 in pre-clinical animals, to elucidate the pharmacokinetic/pharmacodynamic (PK/PD) profiles in mice and PK profiles in monkeys, which can lay the foundation for potential future translation of RBD1016 PK and PD from the pre-clinical stage to the clinic stage. The proposed semi-mechanistic PK/PD model fitted PK and PD data in HBV transgenic mice well and described plasma and liver concentrations in the monkeys well. The simulation results showed that our model has a reasonable predictive ability for Hepatitis B surface antigen (HBsAg) levels after multiple dosing in mice. Further PK and PD data for RBD1016, including clinical data, will assist in refining the model presented here. Our current effort focused on model building for RBD1016, we anticipate that the model could apply to other GalNAc-siRNA drugs.
Collapse
Affiliation(s)
- Qian Li
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taohua Geng
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Haiyan Li
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Shuquan Zheng
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Sara Svedlund
- Ribocure Pharmaceuticals AB, Medicinaregatan 8A, Gothenburg, Sweden
| | - Liming Gan
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
- Ribocure Pharmaceuticals AB, Medicinaregatan 8A, Gothenburg, Sweden
| | - Ann-Charlotte Egnell
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
- Ribocure Pharmaceuticals AB, Medicinaregatan 8A, Gothenburg, Sweden
| | - Shan Gao
- Suzhou Ribo Life Science Co. Ltd., Jiangsu, 215300, China
| | - Rui Chen
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Sandra L, T'jollyn H, Vermeulen A, Ackaert O, Perez‐Ruixo J. Model-based meta-analysis to quantify the effects of short interfering RNA therapeutics on hepatitis B surface antigen turnover in hepatitis B-infected mice. CPT Pharmacometrics Syst Pharmacol 2024; 13:729-742. [PMID: 38522000 PMCID: PMC11098160 DOI: 10.1002/psp4.13129] [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: 10/16/2023] [Revised: 02/24/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
The objective of this study was to compare the efficacy of short interfering RNA therapeutics (siRNAs) in reducing hepatitis B surface antigen (HBsAg) levels in hepatitis B-infected (HBV) mice across multiple siRNA therapeutic classes using model-based meta-analysis (MBMA) techniques. Literature data from 10 studies in HBV-infected mice were pooled, including 13 siRNAs, formulated as liposomal nanoparticles (LNPs) or conjugated to either cholesterol (chol) or N-acetylgalactosamine (GalNAc). Time course of the baseline- and placebo-corrected mean HBsAg profiles were modeled using kinetics of drug effect (KPD) model coupled to an indirect response model (IRM) within a longitudinal non-linear mixed-effects MBMA framework. Single and multiple dose simulations were performed exploring the role of dosing regimens across evaluated siRNA classes. The HBsAg degradation rate (0.72 day-1) was consistent across siRNAs but exhibited a large between-study variability of 31.4% (CV%). The siRNA biophase half-life was dependent on the siRNA class and was highest for GalNAc-siRNAs (21.06 days) and lowest for chol-siRNAs (2.89 days). ID50 estimates were compound-specific and were lowest for chol-siRNAs and highest for GalNAc-siRNAs. Multiple dose simulations suggest GalNAc-siRNAs may require between 4 and 7 times less frequent dosing at higher absolute dose levels compared to LNP-siRNAs and chol-siRNAs, respectively, to reach equipotent HBsAg-lowering effects in HBV mice. In conclusion, non-clinical HBsAg concentration-time data after siRNA administration can be described using the presented KPD-IRM MBMA framework. This framework allows to quantitatively compare the effects of siRNAs on the HBsAg time course and inform dose and regimen selection across siRNA classes. These results may support siRNA development, optimize preclinical study designs, and inform data analysis methodology of future anti-HBV siRNAs; and ultimately, support siRNA model-informed drug development (MIDD) strategies.
Collapse
Affiliation(s)
- Louis Sandra
- Janssen Research and Development, a Johnson & Johnson CompanyBeerseBelgium
- Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical SciencesGhent UniversityGhentBelgium
| | - Huybrecht T'jollyn
- Janssen Research and Development, a Johnson & Johnson CompanyBeerseBelgium
| | - An Vermeulen
- Janssen Research and Development, a Johnson & Johnson CompanyBeerseBelgium
- Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical SciencesGhent UniversityGhentBelgium
| | - Oliver Ackaert
- Janssen Research and Development, a Johnson & Johnson CompanyBeerseBelgium
| | | |
Collapse
|
7
|
Lumen A, Zhang X, Dutta S, Upreti VV. Predicting Clinical Pharmacokinetics/Pharmacodynamics and Impact of Organ Impairment on siRNA-Based Therapeutics Using a Mechanistic Physiologically-Based Pharmacokinetic-Pharmacodynamic Model. Clin Pharmacol Ther 2024; 115:1054-1064. [PMID: 38282246 DOI: 10.1002/cpt.3160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/07/2023] [Indexed: 01/30/2024]
Abstract
Approved and emerging siRNA therapeutics are primarily designed for targeted delivery to liver where the therapeutic gene silencing effects occurs. Impairment of hepatic/renal function and its impact on siRNA pharmacokinetics/pharmacodynamics (PKs/PDs) are yet to be mechanistically evaluated to describe the unanticipated clinical observations for this novel modality. We developed pathophysiologically relevant models for organ impairment within a physiologically-based PK-PD (PBPK-PD) modeling framework focusing on modality-specific mechanistic factors to evaluate impact on siRNA PKs and PDs. PBPK-PD models for two US Food and Drug Administration (FDA) approved siRNAs inclisiran and vutrisiran were developed as case studies leveraging available tissue-specific data and translated to humans. Key determinants of the clinical PK and PD of N-acetylgalactosamine conjugated siRNAs (GalNAc-siRNAs) with varying sequences were also identified to inform effective clinical translation strategies for emerging GalNAc-siRNA candidates. A 30-70% reduction in hepatic asialoglycoprotein receptors concentrations still allowed for sufficient amount of free cytoplasmic siRNA for RISC-loading to produce PD effects comparable in extent and duration to normal liver function. This included severe hepatic impairment for which no clinical data are available. Inclusion of other modality agnostic physiological changes relevant to organ impairment did not alter the findings. Changes in renal physiologies, including changes in GFR across various degrees of impairment, well predicted minimal changes in PD for inclisiran and vutrisiran. This work provides a quantitative mechanistic framework and insights on modality-specific factors that drive clinical translation and patient/disease-related factors that impact specific dosing considerations and clinical outcomes to help accelerate the optimal development of siRNA therapeutics.
Collapse
Affiliation(s)
- Annie Lumen
- Clinical Pharmacology, Modeling, and Simulation, Amgen Inc., South San Francisco, California, USA
| | - Xinwen Zhang
- Clinical Pharmacology, Modeling, and Simulation, Amgen Inc., South San Francisco, California, USA
| | - Sandeep Dutta
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, USA
| | - Vijay V Upreti
- Clinical Pharmacology, Modeling, and Simulation, Amgen Inc., South San Francisco, California, USA
| |
Collapse
|
8
|
Ayyar VS, Song D. Mechanistic Pharmacokinetics and Pharmacodynamics of GalNAc-siRNA: Translational Model Involving Competitive Receptor-Mediated Disposition and RISC-Dependent Gene Silencing Applied to Givosiran. J Pharm Sci 2024; 113:176-190. [PMID: 37871778 DOI: 10.1016/j.xphs.2023.10.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 10/25/2023]
Abstract
Triantennary N-acetyl-D galactosamine (GalNAc)3-conjugated small interfering RNA (siRNA) have majorly advanced the development of RNA-based therapeutics. Chemically stabilized GalNAc-siRNAs exhibit extensive albeit capacity-limited (nonlinear) distribution into hepatocytes with additional complexities in intracellular liver disposition and pharmacology. A mechanism-based pharmacokinetic-pharmacodynamic (PK-PD) model of GalNAc-siRNA was developed to i) quantitate ASGPR-mediated disposition and downstream RNA-induced silencing complex (RISC)-dependent pharmacology following intravenous (IV) and subcutaneous (SC) dosing, ii) assess the kinetics of formed active metabolite, iii) leverage, as an example, published experimental data for givosiran, and iv) demonstrate PK translation across two preclinical species (rat and monkey) with subsequent prediction of human plasma PK. The structural model is based on competition between parent and formed active metabolite for occupancy and uptake via ASGPR into hepatocytes, intracellular sequestration and degradation, and downstream engagement of RNA-induced silencing complex (RISC) governing target mRNA degradation. The model jointly and accurately captured available concentration-time profiles of givosiran and/or AS(N-1)3' givosiran in rat and/or monkey plasma, liver, and/or kidney following givosiran administered both IV and SC. RISC-dependent gene silencing of ALAS1 mRNA was well-characterized. The model estimated an in vivo affinity (KD) value of 27.7 nM for GalNAc-ASGPR and weight-based allometric exponents of -0.27 and -0.24 for SC absorption and intracellular (endolysosomal) degradation rate constants. The model well-predicted reported givosiran plasma PK profiles in humans. PK simulations revealed net-shifts in liver-to-kidney distribution ratios with increasing IV and SC dose. Importantly, decreases in the relative liver uptake efficiency were demonstrated following IV and, to a lesser extent, following SC dosing explained by differential ASGPR occupancy profiles over time.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Clinical Pharmacology & Pharmacometrics, Janssen Research and Development, Spring House, PA, USA.
| | - Dawei Song
- Clinical Pharmacology & Pharmacometrics, Janssen Research and Development, Spring House, PA, USA
| |
Collapse
|
9
|
McGowan A, Gennemark P, Akieh-Pirkanniemi M, Wirman L, Davies N, Elebring M, Tivesten A, Strimfors M, Hölttä M, Söderberg M, Berntsson V, Balas D, Koskinen M, Leino L, Abrahmsén-Alami S. Injectable Biodegradable Silica Depot for Controlled Subcutaneous Delivery of Antisense Oligonucleotides with beyond Monthly Administration. Mol Pharm 2024; 21:143-151. [PMID: 38126776 DOI: 10.1021/acs.molpharmaceut.3c00663] [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: 12/23/2023]
Abstract
Single-stranded antisense oligonucleotides (ASOs) are typically administered subcutaneously once per week or monthly. Less frequent dosing would have strong potential to improve patient convenience and increase adherence and thereby for some diseases result in more optimal therapeutic outcomes. Several technologies are available to provide sustained drug release via subcutaneous (SC) administration. ASOs have a high aqueous solubility and require relatively high doses, which limits the options available substantially. In the present work, we show that an innovative biodegradable, nonporous silica-based matrix provides zero-order release in vivo (rats) for at least 4 weeks for compositions with ASO loads of up to about 100 mg/mL (0.5 mL injection) without any sign of initial burst. This implies that administration beyond once monthly can be feasible. For higher drug loads, substantial burst release was observed during the first week. The concentrations of unconjugated ASO levels in the liver were found to be comparable to corresponding bolus doses. Additionally, infusion using a minipump shows a higher liver exposure than SC bolus administration at the same dose level and, in addition, clear mRNA knockdown and circulating protein reduction comparable to SC bolus dosing, hence suggesting productive liver uptake for a slow-release administration.
Collapse
Affiliation(s)
- Asmaa McGowan
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Peter Gennemark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
- Department of Biomedical Engineering, Linköping University, Linköping 581 83, Sweden
| | | | - Linda Wirman
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Marie Elebring
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Anna Tivesten
- CVRM CMC Projects, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Marie Strimfors
- Bioscience metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Mikko Hölttä
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Magnus Söderberg
- Pathology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Veronica Berntsson
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Daniela Balas
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Mika Koskinen
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Lasse Leino
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Susanna Abrahmsén-Alami
- Sustainable Innovation & Transformational Excellence, Pharmaceutical Technology & Development, Operations, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| |
Collapse
|
10
|
An G. Pharmacokinetics and Pharmacodynamics of GalNAc-Conjugated siRNAs. J Clin Pharmacol 2024; 64:45-57. [PMID: 37589246 DOI: 10.1002/jcph.2337] [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: 06/07/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Small interfering RNAs (siRNAs) represent a new class of drugs with tremendous potential for battling previously "undruggable" diseases. After nearly 2 decades of efforts in addressing the problems of the poor drug profile of naked unmodified siRNAs, this new modality has finally come to fruition, with 5 agents (patisiran, givosiran, lumasiran, inclisiran, and vutrisiran) being approved since 2018, and with many others in the different phases of clinical development. Unlike small-molecule drugs and protein therapeutics, siRNAs have different sizes, distinct mechanisms of action, differing physicochemical and pharmacological properties, and, accordingly, a unique pharmacokinetic/pharmacodynamic (PK/PD) relationship. To support the continuous development of siRNAs, it is important to have a thorough and deep understanding of the PK/PD and clinical pharmacology related features of siRNAs. As most of the current siRNA products are conjugated by N-acetylgalactosamine (GalNAc), this review focuses on the PK/PD relationships and clinical pharmacology of GalNAc-conjugated siRNAs, including their absorption, distribution, metabolism, excretion (ADME) properties, PK/PD models, drug-drug interactions, clinical pharmacology in special populations, and safety evaluation. In addition, necessary background information related to the development of siRNAs as a therapeutic modality, including the mechanisms of action, the advantages of siRNAs, the problems of naked siRNAs, as well as the strategies used to enhance the clinical utility of siRNAs, have also been covered. The goal of this review is to serve as a "primer" on siRNA PK/PD, and I hope the readers, especially those who have a limited background on siRNA therapeutics, will have a fundamental understanding of siRNA PK/PD and clinical pharmacology after reading this review.
Collapse
Affiliation(s)
- Guohua An
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
11
|
Sten S, Cardilin T, Antonsson M, Gennemark P. Plasma Pharmacokinetics of N-Acetylgalactosamine-Conjugated Small-Interfering Ribonucleic Acids (GalNAc-Conjugated siRNAs). Clin Pharmacokinet 2023; 62:1661-1672. [PMID: 37824025 PMCID: PMC10684612 DOI: 10.1007/s40262-023-01314-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2023] [Indexed: 10/13/2023]
Abstract
Small-interfering ribonucleic acids (siRNAs) with N-acetylgalactosamine (GalNAc) conjugation for improved liver uptake represent an emerging class of drugs that modulate liver-expressed therapeutic targets. The pharmacokinetics of GalNAc-siRNAs are characterized by a rapid distribution from plasma to tissue (hours) and a long terminal plasma half-life, analyzed in the form of the antisense strand, driven by redistribution from tissue (weeks). Understanding how clinical pharmacokinetics relate to the dose and type of siRNA chemical stabilizing method used is critical, e.g., to design studies, to investigate safety windows, and to predict the pharmacokinetics of new preclinical assets. To this end, we collected and analyzed pharmacokinetic data from the literature regarding nine GalNAc-siRNAs. Based on this analysis, we showed that the clinical plasma pharmacokinetics of GalNAc-siRNAs are approximately dose proportional and similar between chemical stabilizing methods. This holds for both the area under the concentration-time curve (AUC) and the maximum plasma concentration (Cmax). Corresponding rat and monkey pharmacokinetic data for a subset of the nine GalNAc-siRNAs show dose-proportional Cmax, supra-dose-proportional AUC, and similar pharmacokinetics between chemical stabilizing methods. Together, the animal and human pharmacokinetic data indicate that plasma clearance divided by bioavailability follows allometric principles and scales between species with an exponent of 0.75. Finally, the clinical plasma concentration-time profiles can be empirically described by standard one-compartment kinetics with first-order absorption up to 24 h after subcutaneous dosing, and by three-compartment kinetics with first-order absorption in general. To describe the system more mechanistically, we report a corrected and unambiguously defined version of a previously published physiologically based pharmacokinetic model.
Collapse
Affiliation(s)
- Sebastian Sten
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Tim Cardilin
- Fraunhofer-Chalmers Research Centre for Industrial Mathematics, Chalmers Science Park, 41288, Gothenburg, Sweden
| | - Madeleine Antonsson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Peter Gennemark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden.
| |
Collapse
|