13 results on '"GalNAc-siRNA"'
Search Results
2. Preclinical and Clinical Pharmacokinetics of JNJ‐75220795, an siRNA Therapeutic Targeting PNPLA3, for Metabolic Dysfunction‐Associated Steatohepatitis.
- Author
-
Jeon, Jae Yoon, Ayyar, Vivaswath S., Ouchi, Shohei, Fabbrini, Elisa, Koshkina, Anastasiya, Prusakiewicz, Jeffery J., Dallas, Jed, Yang, Txheng, Jian, Wenying, Kang, Lijuan, Cofsky, Korin, Rady, Brian, Tamamura, Ryo, Saito, Yuki, Yamashita, Aimi, Vaughan, Tamisha, Wendel, Susan, Makimura, Hideo, Csonka, Dénes, and Goyal, Navin
- Subjects
- *
RNA , *PHARMACOKINETICS , *FATTY liver , *RATS , *SMALL interfering RNA - Abstract
JNJ‐75220795 or ARO‐PNPLA3 is an investigational small interfering ribonucleic acid agent conjugated with N‐acetyl‐d‐galactosamine that targets the
PNPLA3 gene, currently being developed for metabolic dysfunction‐associated steatohepatitis (MASH). This study evaluated the pharmacokinetics (PK) profile of single subcutaneous doses of JNJ‐75220795 in preclinical species as well as in human subjects with homozygous or heterozygousPNPLA3 I148M mutation in two phase 1 studies—a first‐in‐human study in the United States and a first‐in‐Japanese study in Japan. Preclinical PK in rats and non‐human primates (NHP) showed a rapid systemic absorption and elimination following single subcutaneous doses. JNJ‐75220795 was predominantly distributed to the liver with peak liver concentrations reached at 4 h and still detectable at 672 and 336 h in rats and NHPs, respectively, with an apparent liver‐to‐plasma area under the curve (AUC) ratio of 2800 in rats. Consistent with the preclinical findings, clinical PK showed rapid systemic absorption and clearance in humans with median peak concentrations at 3.0–9.0 h and mean short half‐life of 3.4–6.2 h. Plasma PK exposure parameters including Cmax and AUC increased approximately dose proportionally. Kidney had the second highest tissue exposure following the liver in rats. Renal excretion was a significant but minor elimination pathway as approximately 15%–25% of the administered dose was recovered in urine. Based on the overall data, JNJ‐75220795 was primarily localized to the liver and exhibited sustained hepatic exposures, which confer prolonged pharmacodynamic effects in the target organ. The favorable PK profiles of single subcutaneous doses observed in the phase 1 studies support continued clinical development of JNJ‐75220795 for the treatment of MASH. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
3. RNAi targeting LMAN1-MCFD2 complex promotes anticoagulation in mice: RNAi targeting LMAN1-MCFD2 complex promotes anticoagulation in mice: S. Ma et al.
- Author
-
Ma, Siqian, Liu, Boyan, Du, Hong, Yang, Fei, Han, Jingjing, Huang, Xinqi, Zhang, Minyang, Ji, Shundong, and Jiang, Miao
- Abstract
Combined deficiency of coagulation factor V (FV) and factor VIII (FVIII) is a rare bleeding disease caused by variants in either lectin mannose binding 1 (LMAN1) or multiple coagulation factor deficiency 2 (MCFD2) gene. Reducing the level of FVIII by inhibiting the LMAN1-MCFD2 complex may become a new anticoagulant approach. We aimed to find a new therapeutic option for anticoagulation by RNA interference (RNAi) targeting LMAN1 and MCFD2. siRNA sequences with cross-homology between mice and humans were designed based on LMAN1 or MCFD2 transcripts in NCBI and were screened with the Dual-Luciferase reporter assay. The optimal siRNAs were chemically modified and conjugated with three N-acetylgalactosamine molecules (GalNAc-siRNA), promoting their targeted delivery to the liver. The expression of LMAN1 and MCFD2 in cell lines or mice was examined by RT-qPCR and western blotting. For the mice administered with siRNA, we assessed their coagulation function by measuring APTT and the activity of FVIII factor. After administration, siRNAs GalNAc-LMAN1 and GalNAc-MCFD2 demonstrated effective and persistent LMAN1 and MCFD2 inhibition. 7 days after injection of 3mg/kg GalNAc-LMAN1, the LMAN1 mRNA levels reduced to 19.97% ± 3.78%. MCFD2 mRNA levels reduced to 32.22% ± 13.14% with injection of 3mg/kg GalNAc-MCFD2. After repeated administration, APTT was prolonged and the FVIII activity was remarkably decreased. The tail bleeding test of mice showed that the amount of bleeding in the treated group did not significantly increase compared with the control group. Our study confirms that therapy with RNAi targeting LMAN1-MCFD2 complex is effective and can be considered a viable option for anticoagulation drugs. However, the benefits and potential risk of bleeding in thrombophilic mice model needs to be evaluated. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Targeting Liver Xor by GalNAc-siRNA Is an Effective Strategy for Hyperuricemia Therapy.
- Author
-
Sun, Huiyan, Wang, Xinxia, Li, Yongqiang, Shen, Yingzhi, Zhang, Lin, Xu, Yingjie, Liu, Junling, and Fan, Xuemei
- Subjects
- *
MICE , *URIC acid , *KNOCKOUT mice , *CARDIOVASCULAR diseases , *HYPERURICEMIA - Abstract
Hyperuricemia, i.e., increased plasma uric acid concentration, is a common problem in clinical practice, leading to gout or nephrolithiasis, and is associated with other disorders, such as metabolic syndrome, cardiovascular disease, and chronic renal disease. Xanthine oxidoreductase (XOR) is a critical rate-limiting enzyme involved in uric acid synthesis and a promising target for hyperuricemia therapy. However, XOR inhibitors currently face clinical problems such as a short half-life and side effects. Here, we found that specifically targeting liver Xor with GalNAc-siRNAs had a good therapeutic effect on hyperuricemia. First, siRNAs were designed to target various sites in the homologous region between Homo sapiens and Mus musculus Xor mRNA and were screened in primary mouse hepatocytes. Then, the siRNAs were modified to increase their stability in vivo and conjugated with GalNAc for liver-specific delivery. The effects of GalNAc-siRNAs were evaluated in three hyperuricemia mouse models, including potassium oxonate and hypoxanthine administration in WT and humanized XDH mice and Uox knockout mice. Febuxostat, a specific XOR inhibitor used for hyperuricemia treatment, was used as a positive control. Targeting liver Xor with GalNAc-siRNAs by subcutaneous administration reduced plasma uric acid levels, uric acid accumulation in the kidney, renal inflammation, and fibrosis, thereby alleviating kidney damage in hyperuricemia mouse models without hepatoxicity. The results demonstrated that targeting liver Xor with GalNAc-siRNAs was a promising strategy for hyperuricemia therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Efficacy of GalNAc C3 siRNAs in factor H-deficient mice with C3 glomerulopathy.
- Author
-
Zanchi, Cristina, Locatelli, Monica, Cerullo, Domenico, Aumiller, Verena, Corna, Daniela, Rottoli, Daniela, Schubert, Steffen, Noris, Marina, Tomasoni, Susanna, Remuzzi, Giuseppe, Zoja, Carlamaria, and Benigni, Ariela
- Subjects
- *
MICE , *SMALL interfering RNA , *COMPLEMENT (Immunology) , *COMPLEMENT factor H - Abstract
Complement alternative pathway (AP) dysregulation drives C3 glomerulopathy (C3G), a rare renal disorder characterized by glomerular C3 deposition and glomerular damage, for which no effective treatments are available. Blockade of complement C3 is emerging as a viable therapeutic option. In an earlier study we showed that SLN500, a small interfering RNA targeting liver C3 synthesis, was able to limit AP dysregulation and glomerular C3d deposits in mice with partial factor H (FH) deficiency (Cfh +/- mice). Here, we assessed the pharmacological effects of SLN501 – an optimized SLN500 version – in mice with complete FH deficiency (Cfh -/- mice) that exhibit a more severe C3G phenotype. SLN501 effectively prevented liver C3 synthesis, thus limiting AP dysregulation, glomerular C3d deposits and the development of ultrastructural alterations. These data provide firm evidence of the use of siRNA-mediated liver C3 gene silencing as a potential therapy for treating C3G patients with either partial or complete FH loss of function. • A GalNAc anti-C3 siRNA, SLN501, silences liver C3 synthesis in Cfh -/- mice. • SLN501 limits fluid-phase complement dysregulation in Cfh -/- mice. • SLN501 reduces glomerular C3d and mesangial electron-dense deposits in Cfh -/- mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Hepatocyte-targeted siTAZ therapy lowers liver fibrosis in NASH diet-fed chimeric mice with hepatocyte-humanized livers
- Author
-
Xiaobo Wang, Mary P. Moore, Hongxue Shi, Yoshinari Miyata, Sara K. Donnelly, Daniel R. Radiloff, and Ira Tabas
- Subjects
hepatocyte-humanized mice ,MASH ,liver fibrosis ,WWTR1 ,GalNAc-siRNA ,NASH/MASH therapy, NASH, TAZ, Indian hedgehog ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Nonalcoholic steatohepatitis (NASH) is emerging as the most common cause of liver disease. Although many studies in mouse NASH models have suggested therapies, translation to humans is poor, with no approved drugs for NASH. One explanation may lie in differences between mouse and human hepatocytes. We used NASH diet-fed chimeric mice reconstituted with human hepatocytes (hu-liver mice) to test a mechanism-based hepatocyte-targeted small interfering RNA (siRNA), GalNAc-siTaz, shown previously to block the progression to fibrotic NASH in mice. Following ablation of endogenous hepatocytes, male mice were reconstituted with human hepatocytes from a single donor with the rs738409-C/G PNPLA3 risk variant, resulting in ∼95% human hepatocyte reconstitution. The mice were then fed a high-fat choline-deficient l-amino acid–defined diet for 6 weeks to induce NASH, followed by six weekly injections of GalNAc-siTAZ to silence hepatocyte-TAZ or control GalNAc-siRNA (GalNAc-control) while still on the NASH diet. GalNAc-siTAZ lowered human hepatic TAZ and IHH, a TAZ target that promotes NASH fibrosis. Most important, GalNAc-siTAZ decreased liver inflammation, hepatocellular injury, hepatic fibrosis, and profibrogenic mediator expression versus GalNAc-control, indicating that GalNAc-siTAZ decreased the progression of NASH in mice reconstituted with human hepatocytes. In conclusion, silencing TAZ in human hepatocytes suppresses liver fibrosis in a hu-liver model of NASH.
- Published
- 2023
- Full Text
- View/download PDF
7. Liver factor B silencing to cure C3 glomerulopathy: Evidence from a mouse model of complement dysregulation.
- Author
-
Zanchi, Cristina, Locatelli, Monica, Corna, Daniela, Cerullo, Domenico, Fishilevich, Elane, Desai, Dhruv, Rottoli, Daniela, Donadelli, Roberta, Noris, Marina, Zoja, Carlamaria, Remuzzi, Giuseppe, and Benigni, Ariela
- Subjects
- *
MICE , *COMPLEMENT factor H , *LABORATORY mice , *ANIMAL disease models , *GENE expression , *PARIETAL cells , *LIVER - Abstract
Uncontrolled activation of the alternative pathway (AP) of complement, due to genetic and/or acquired defects, plays a primary pathogenetic role in C3 glomerulopathy (C3G), a rare and heterogeneous disease characterised by predominant C3 fragment deposition within the glomerulus, as well as glomerular damage. There are currently no approved disease-specific treatments for C3G, but new drugs that directly counteract AP dysregulation, targeting components of the pathway, have opened promising new perspectives for managing the disease. Complement factor B (FB), which is primarily synthesised by hepatocytes, is a key component of the AP, as it drives the central amplification loop of the complement system. In this study we used a GalNAc (N-Acetylgalactosamine)-conjugated siRNA to selectively target and suppress liver FB expression in two mouse models characterised by the complete (Cfh -/- mice) or partial (Cfh +/- ) loss of function of complement factor H (FH). Homozygous deletion of FH induced a severe C3G phenotype, with strong dysregulation of the AP of complement, glomerular C3 deposition and almost complete C3 consumption. Mice with a heterozygous deletion of FH had intermediate C3 levels and exhibited slower disease progression, resembling human C3G more closely. Here we showed that FB siRNA treatment did not improve serum C3 levels, nor limit glomerular C3 deposition in Cfh -/- mice, while it did normalise circulating C3 levels, reduce glomerular C3 deposits, and limit mesangial electron-dense deposits in Cfh +/- mice. The present data provide important insights into the potential benefits and limitations of FB-targeted inhibition strategies and suggest RNA interference-mediated FB silencing in the liver as a possible therapeutic approach for treating C3G patients with FH haploinsufficiency. • A GalNAc FB siRNA silences liver FB gene expression in Cfh -/- and Cfh +/- mice. • FB siRNA reduces circulating FB levels in both the Cfh -/- and Cfh +/- mice. • Serum C3 levels are normalized by treatment with FB siRNA only in Cfh +/- mice. • FB siRNA reduces glomerular C3 and mesangial electron-dense deposits in Cfh +/- mice. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. The therapeutic prospects of N-acetylgalactosamine-siRNA conjugates
- Author
-
Lei Zhang, Yayu Liang, Guohui Liang, Zhili Tian, Yue Zhang, Zhihui Liu, and Xinying Ji
- Subjects
RNAi ,GalNAc-siRNA ,delivery system ,ASGPR ,LNPs ,Therapeutics. Pharmacology ,RM1-950 - Abstract
RNA interference has become increasingly used for genetic therapy following the rapid development of oligonucleotide drugs. Significant progress has been made in its delivery system and implementation in the treatment of target organs. After a brief introduction of RNA interference technology and siRNA, the efficiency and stability of GalNAc-siRNA conjugates are highlighted since several oligonucleotide drugs of GalNAc have been approved for clinical use in recent years. The structure and features of GalNAc-siRNA conjugates are studied and the clinical efficiency and limitations of oligonucleotide-based drugs are summarized and investigated. Furthermore, another delivery system, lipid nanoparticles, that confer many advantages, is concluded, includ-ing stability and mass production, compared with GalNAc-siRNA conjugates. Importantly, developing new approaches for the use of oligonucleotide drugs brings hope to genetic therapy.
- Published
- 2022
- Full Text
- View/download PDF
9. Less Is More: Novel Hepatocyte-Targeted siRNA Conjugates for Treatment of Liver-Related Disorders
- Author
-
Adrien Weingärtner, Lucas Bethge, Lisa Weiss, Maria Sternberger, and Marie Wikstrom Lindholm
- Subjects
siRNA-conjugate ,GalNAc-siRNA ,serinol-GalNAc ,asialoglycoprotein ,GalNAc-cluster ,Therapeutics. Pharmacology ,RM1-950 - Abstract
N-acetyl-galactosamine (GalNAc) conjugation enhances liver specificity for therapeutic oligonucleotides. Here we report on a novel design with improved activity and stability compared with a triantennary design. We applied a versatile monovalent serinol-GalNAc conjugation strategy. First, 1–4 serial serinol-linked GalNAc units were conjugated to terminal positions of small interfering RNA (siRNA) molecules. In primary hepatocytes, 5′ antisense GalNAc conjugates were inactive, whereas 3′ antisense and 3′ or 5′ sense conjugates displayed low activity for single GalNAc units, while 2–4 serial GalNAc conjugates were all equally potent. In mice, 5′ sense conjugates with 2–4 serial GalNAc units were all as potent as a triantennary GalNAc control (1 mg/kg). Second, increased spacing between two serial 5′ sense-conjugated GalNAc units did not affect in vitro activity. Finally, two single GalNAc units were positioned at opposite ends of the sense strand. A single dose (0.3 mg/kg) of this novel conjugate in mice showed a 3-fold reduction of serum target protein level at day 7 and 4-fold lower serum level at day 27, relative to an equimolar dose of a triantennary GalNAc conjugate of the same siRNA. Improved tritosome stability (by liquid chromatography-mass spectrometry [LC-MS] analysis) can at least partially explain the increased activity and duration of action for the novel GalNAc conjugate.
- Published
- 2020
- Full Text
- View/download PDF
10. Knockdown of liver-derived factor XII by GalNAc-siRNA ALN-F12 prevents thrombosis in mice without impacting hemostatic function.
- Author
-
Liu, Jingxuan, Cooley, Brian C., Akinc, Akin, Butler, James, and Borodovsky, Anna
- Subjects
- *
THROMBOSIS , *VENOUS thrombosis , *BLOOD coagulation factors , *FERRIC chloride , *SAPHENOUS vein - Abstract
Plasma coagulation Factor XII (FXII) plays a crucial role in contact activation, ultimately regulating both the kinin-kallikrein system and the intrinsic pathway of coagulation. A growing body of evidence suggests that inhibition of FXII can prevent thrombosis. Given FXII does not appear to modulate hemostasis, targeting FXII is a promising strategy for the prevention of pathological thrombus formation without the hemostatic risks typically associated with anticoagulants. To this end, a subcutaneously administered investigational RNAi therapeutic targeting liver F12 mRNA (ALN-F12) was developed. To investigate the thrombo-protective and hemostatic effects of FXII reduction by ALN-F12 in rodent thrombosis and hemostasis models. A single dose of ALN-F12 was subcutaneously administered to C57Bl/6 mice. After reaching steady state FXII reduction, the impact on thrombosis (ferric chloride arterial thrombosis and electrolytic injury induced venous thrombosis models) and hemostasis (saphenous vein injury and tail tip transection bleeding models) was evaluated. Administration of ALN-F12 resulted in dose-dependent reductions of both liver F12 mRNA and plasma FXII protein. In mice, ALN-F12 led to dose-dependent reductions in platelet and fibrin accumulation in the venous electrolytic-injury model and in the time to occlusion in the ferric chloride arterial thrombosis model. At 10 mg/kg ALN-F12, the top dose level evaluated, this resulted in >95% reduction of FXII and ~10 fold reduction in fibrin deposition. Finally, hemostasis models showed that >95% reduction of FXII had no impact on bleeding time or blood loss. Our findings support that reduction of plasma Factor XII by ALN-F12 provided thrombo-protective effects with no increased bleeding risk in rodent models of thrombosis and hemostasis. • Plasma coagulation factor FXII is a potential target for thromboprophylaxis. • GalNAc-siRNA ALN-F12 knocks down liver F12 and reduces plasma FXII levels. • ALN-F12 treatment prevents thrombosis in mouse models. • F12 knock down by ALN-F12 does not impact hemostasis in mice. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
11. Evaluation of GalNAc-siRNA Conjugate Activity in Pre-clinical Animal Models with Reduced Asialoglycoprotein Receptor Expression
- Author
-
Theo J.C. Van Berkel, Kun Qian, Muthiah Manoharan, Kallanthottathil G. Rajeev, Vasant Jadhav, Klaus Charisse, Jayaprakash K. Nair, James Butler, Tracy Zimmermann, Amy Chan, Jennifer L. S. Willoughby, Alfica Sehgal, Kristina Yucius, Tim Racie, Tuyen Nguyen, Svetlana Shulga-Morskaya, and Martin Maier
- Subjects
Liver Cirrhosis ,0301 basic medicine ,Acetylgalactosamine ,Receptor expression ,Drug Evaluation, Preclinical ,Asialoglycoprotein Receptor ,GalNAc-siRNA ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,RNA interference ,Pharmacokinetics ,Drug Discovery ,Genetics ,Animals ,Humans ,Gene Silencing ,RNA, Small Interfering ,Receptor ,Molecular Biology ,Mice, Knockout ,Pharmacology ,Drug Carriers ,Chemistry ,Ligand (biochemistry) ,Cell biology ,carbohydrates (lipids) ,Disease Models, Animal ,Protein Subunits ,030104 developmental biology ,Gene Expression Regulation ,Targeted drug delivery ,Biochemistry ,Hepatocytes ,Molecular Medicine ,Female ,Original Article ,lipids (amino acids, peptides, and proteins) ,Asialoglycoprotein receptor ,Function (biology) ,Conjugate - Abstract
The hepatocyte-specific asialoglycoprotein receptor (ASGPR) is an ideal candidate for targeted drug delivery to the liver due to its high capacity for substrate clearance from circulation together with its well-conserved expression and function across species. The development of GalNAc-siRNA conjugates, in which a synthetic triantennary N-acetylgalactosamine-based ligand is conjugated to chemically modified siRNA, has enabled efficient, ASGPR-mediated delivery to hepatocytes. To investigate the potential impact of variations in receptor expression on the efficiency of GalNAc-siRNA conjugate delivery, we evaluated the pharmacokinetics and pharmacodynamics of GalNAc-siRNA conjugates in multiple pre-clinical models with reduced receptor expression. Despite greater than 50% reduction in ASGPR levels, GalNAc conjugate activity was retained, suggesting that the remaining receptor capacity was sufficient to mediate efficient uptake of potent GalNAc-siRNAs at pharmacologically relevant dose levels. Collectively, our data support a broad application of the GalNAc-siRNA technology for hepatic targeting, including disease states where ASGPR expression may be reduced., Willoughby and colleagues show that GalNAc-siRNA conjugate uptake is specifically mediated through the hepatic expressed asialoglycoprotein receptor and that potent conjugates are capable of robust gene silencing in reduced receptor settings. These data combined support the therapeutic potential in disease(s) where receptor levels may be reduced due to liver injury.
- Published
- 2018
- Full Text
- View/download PDF
12. An investigational RNAi therapeutic targeting Factor XII (ALN-F12) for the treatment of hereditary angioedema.
- Author
-
Liu J, Qin J, Borodovsky A, Racie T, Castoreno A, Schlegel M, Maier MA, Zimmerman T, Fitzgerald K, Butler J, and Akinc A
- Subjects
- Animals, Capillary Permeability drug effects, Complement C1 Inhibitor Protein genetics, Factor XII analysis, Female, Humans, Kininogens metabolism, Macaca fascicularis, Mice, Mice, Inbred C57BL, RNA Interference, Rats, Angioedemas, Hereditary drug therapy, Bradykinin biosynthesis, Factor XII genetics, RNA, Small Interfering genetics, RNA, Small Interfering therapeutic use
- Abstract
Hereditary angioedema (HAE) is a genetic disorder mostly caused by mutations in the C1 esterase inhibitor gene (C1INH) that results in poor control of contact pathway activation and excess bradykinin generation. Bradykinin increases vascular permeability and is ultimately responsible for the episodes of swelling characteristic of HAE. We hypothesized that the use of RNA interference (RNAi) to reduce plasma Factor XII (FXII), which initiates the contact pathway signaling cascade, would reduce contact pathway activation and prevent excessive bradykinin generation. A subcutaneously administered GalNAc-conjugated small-interfering RNA (siRNA) targeting F12 mRNA (ALN-F12) was developed, and potency was evaluated in mice, rats, and cynomolgus monkeys. The effect of FXII reduction by ALN-F12 administration was evaluated in two different vascular leakage mouse models. An ex vivo assay was developed to evaluate the correlation between human plasma FXII levels and high-molecular weight kininogen (HK) cleavage. A single subcutaneous dose of ALN-F12 led to potent, dose-dependent reduction of plasma FXII in mice, rats, and NHP. In cynomolgus monkeys, a single subcutaneous dose of ALN-F12 at 3 mg/kg resulted in >85% reduction of plasma FXII. Administration of ALN-F12 resulted in dose-dependent reduction of vascular permeability in two different mouse models of bradykinin-driven vascular leakage, demonstrating that RNAi-mediated reduction of FXII can potentially mitigate excess bradykinin stimulation. Lastly, ex vivo human plasma HK cleavage assay indicated FXII-dependent bradykinin generation. Together, these data suggest that RNAi-mediated knockdown of FXII by ALN-F12 is a potentially promising approach for the prophylactic treatment of HAE., (© 2019 Liu et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2019
- Full Text
- View/download PDF
13. Clinical Proof of Concept for a Novel Hepatocyte-Targeting GalNAc-siRNA Conjugate.
- Author
-
Zimmermann TS, Karsten V, Chan A, Chiesa J, Boyce M, Bettencourt BR, Hutabarat R, Nochur S, Vaishnaw A, and Gollob J
- Subjects
- Adult, Asialoglycoprotein Receptor genetics, Asialoglycoprotein Receptor metabolism, Drug Monitoring, Female, Gene Silencing, Healthy Volunteers, Humans, Male, Middle Aged, Prealbumin genetics, RNA, Small Interfering chemistry, RNA, Small Interfering pharmacology, Young Adult, Acetylgalactosamine chemistry, Hepatocytes metabolism, RNA, Small Interfering genetics, RNA, Small Interfering therapeutic use
- Abstract
Advancement of RNAi-based therapeutics depends on effective delivery to the site of protein synthesis. Although intravenously administered, multi-component delivery vehicles have enabled small interfering RNA (siRNA) delivery and progression into clinical development, advances of single-component, systemic siRNA delivery have been challenging. In pre-clinical models, attachment of a triantennary N-acetylgalactosamine (GalNAc) ligand to an siRNA mediates hepatocyte uptake via the asialoglycoprotein receptor enabling RNAi-mediated gene silencing. In this phase 1 study, we assessed translation of this delivery approach by evaluating the safety, tolerability, pharmacokinetics, and pharmacodynamics of a GalNAc-siRNA conjugate, revusiran, targeting transthyretin (TTR). Subjects received a placebo or ascending doses of revusiran subcutaneously ranging from 1.25-10 mg/kg in the single and 2.5-10 mg/kg in the multiple ascending dose phases. Revusiran was generally well tolerated, with transient, mild to moderate injection site reactions the most common treatment-emergent adverse events. Doses of 2.5-10 mg/kg revusiran elicited a significant reduction of serum TTR versus the placebo (p < 0.01), with mean TTR reductions of approximately 90% observed with multiple dosing. These results demonstrate translation of this novel delivery platform, enabling clinical development of subcutaneously administered GalNAc-siRNAs for liver-based diseases., (Copyright © 2017 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.