Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 482
Filter
1.
Nucleic Acids Res ; 52(10): 5423-5437, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38742636

ABSTRACT

Oral delivery is the most widely used and convenient route of administration of medicine. However, oral administration of hydrophilic macromolecules is commonly limited by low intestinal permeability and pre-systemic degradation in the gastrointestinal (GI) tract. Overcoming some of these challenges allowed emergence of oral dosage forms of peptide-based drugs in clinical settings. Antisense oligonucleotides (ASOs) have also been investigated for oral administration but despite the recent progress, the bioavailability remains low. Given the advancement with highly potent and durable trivalent N-acetylgalactosamine (GalNAc)-conjugated small interfering RNAs (siRNAs) via subcutaneous (s.c.) injection, we explored their activities after oral administration. We report robust RNA interference (RNAi) activity of orally administrated GalNAc-siRNAs co-formulated with permeation enhancers (PEs) in rodents and non-human primates (NHPs). The relative bioavailability calculated from NHP liver exposure was <2.0% despite minimal enzymatic degradation in the GI. To investigate the impact of oligonucleotide size on oral delivery, highly specific GalNAc-conjugated single-stranded oligonucleotides known as REVERSIRs with different lengths were employed and their activities for reversal of RNAi effect were monitored. Our data suggests that intestinal permeability is highly influenced by the size of oligonucleotides. Further improvements in the potency of siRNA and PE could make oral delivery of GalNAc-siRNAs as a practical solution.


Subject(s)
Acetylgalactosamine , RNA, Small Interfering , Animals , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Administration, Oral , Mice , Rats , RNA Interference , Male , Biological Availability , Humans , Rats, Sprague-Dawley , Macaca fascicularis , Liver/metabolism , Macaca mulatta
2.
Drug Deliv ; 29(1): 574-587, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35156491

ABSTRACT

A novel hyaluronic acid (HA)-modified hybrid nanocomplex HA-SeSe-COOH/siR-93C@PAMAM, which could efficiently deliver siRNA into tumor cells via a redox-mediated intracellular disassembly, was constructed for enhanced antitumor efficacy. Thereinto, siR-93C (siRNA) and positive PAMAM were firstly mixed into the electrostatic nano-intermediate, and then diselenide bond (-SeSe-)-modified HA was coved to shield excessive positive charges. This hybrid nanocomplex displayed uniform dynamic sizes, high stability, controlled zeta potential and narrow PDI distribution. Moreover, the -SeSe- linkage displayed GSH/ROS dual responsive properties, improving intracellular trafficking of siRNA. In vitro assays in A549 cell line presented that HA-SeSe-COOH/siR-93C@PAMAM has low cytotoxicity, rapid lysosomal escape and significant transfection efficiency; besides, an efficient proliferation inhibition ability and enhanced apoptosis. Furthermore, in animal studies, this negative-surfaced hybrid nanocomplex showed a prolonged circulation in blood and improved inhibition of tumor growth. All these results verified our hypothesis in this study that diselenide bonds-modified HA could promote not only stability and safety of nanoparticles in vivo but also intracellular behavior of siRNA via redox-dual sensitive properties; furthermore, this hybrid nanocomplex provided a visible potential approach for siRNA delivery in the antitumor field.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Nanoparticles/chemistry , RNA, Small Interfering/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Dendrimers/chemistry , Drug Carriers/chemistry , Drug Liberation , Hyaluronic Acid/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nylons/chemistry , Oxidation-Reduction , Particle Size , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , Surface Properties
3.
J Clin Lipidol ; 16(2): 208-219, 2022.
Article in English | MEDLINE | ID: mdl-35168913

ABSTRACT

BACKGROUND: Inclisiran, a small interfering RNA molecule, reduces low-density lipoprotein cholesterol (LDL-C) by inhibiting production of proprotein convertase subtilisin/kexin type 9 (PCSK9) in the liver. OBJECTIVE: To investigate the pharmacokinetics, pharmacodynamics, and safety of inclisiran in patients with mild or moderate hepatic impairment (HI) vs participants with normal hepatic function (NHF). METHODS: In this single-center, open-label, parallel-group study, patients with mild (Child-Pugh A) or moderate (Child-Pugh B) HI and with NHF, matched by age, body mass index, sex, and race (if possible), received a single subcutaneous therapeutic dose of inclisiran (300 mg). Pharmacokinetic profiles, pharmacodynamic endpoints (PCSK9 and LDL-C), and safety were assessed. RESULTS: Twenty-eight participants completed the study (mild HI: n = 10; moderate HI: n = 6; NHF: n = 12). Inclisiran achieved maximum plasma concentration at 4-6 h and was undetectable in plasma at 48 h in most participants, irrespective of liver function. Inclisiran exposure was 1.24-fold higher in the mild HI vs NHF groups (90% confidence interval [CI] 1.01-1.53) and 2.03-fold higher in the moderate HI vs NHF groups (90% CI 1.60-2.58). LDL-C and PCSK9 plasma levels decreased from baseline up to the last assessment on Day 60 in all groups, with a similar response in NHF and mild HI groups but a less pronounced and more varied decrease in the moderate HI group. Inclisiran was generally safe and well tolerated. CONCLUSION: The pharmacokinetic exposure of inclisiran increased by up to two fold in patients with moderate HI compared with those with NHF, while pharmacodynamic effects remained relatively unchanged. Inclisiran is generally safe and well tolerated in patients with mild or moderate HI, with no dose adjustment needed. However, a larger, long-term clinical trial would help to further evaluate the long-term safety profile of inclisiran in patients with liver disease.


Subject(s)
Liver Diseases , RNA, Small Interfering , Cholesterol, LDL , Humans , Liver Diseases/drug therapy , Proprotein Convertase 9/genetics , RNA, Small Interfering/adverse effects , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
4.
Biotechnol Bioeng ; 119(2): 482-492, 2022 02.
Article in English | MEDLINE | ID: mdl-34761810

ABSTRACT

As one of the gene therapies, RNA interference (RNAi) effectively suppresses only specific genes, targeting various diseases in which they are involved. For the successful process of RNAi, efficient and safe delivery of small RNAs, including small interfering RNA and short hairpin RNA, is essential. Herein, an S-R11 fusion peptide, SPACE peptide conjugated with poly-arginine, was introduced to deliver small RNAs into immune cells that are difficult to transfect. This S-R11 peptide stably formed a spontaneous self-assembling nanocomplex through electrostatic attraction and hydrogen bonding with small RNAs. The nanocomplex showed about 5.3-fold better permeation efficiency than the conventional Lipofectamine™ 2000 for RAW 264.7 macrophage cells. Moreover, it induced about 66.2% silencing effect of the target gene in the cells activated with polyinosinic:polycytidylic acid (poly (I:C)). In addition, the cell viability of fusion peptide was ensured even in a concentration range exceeding the concentration used in the nanocomplex. Based on these results, it is expected that the nanocomplex in this study can be used as a new gene delivery system that can overcome the challenge of gene therapies to immune cells.


Subject(s)
Drug Delivery Systems/methods , Nanostructures/chemistry , Peptides/chemistry , RNA Interference , RNA, Small Interfering , Animals , Genetic Therapy , Macrophages , Mice , RAW 264.7 Cells , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics
5.
Int J Mol Sci ; 22(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34948105

ABSTRACT

Cell penetrating peptides (CPPs) are peptides that can directly adapt to cell membranes and then permeate into cells. CPPs are usually covalently linked to the surface of nanocarriers to endow their permeability to the whole system. However, hybrids with lipids or polymers make the metabolism much more sophisticated and even more difficult to determine. In this study, we present a continuous sequence of 18 amino acids (FFAARTMIWY(d-P)GAWYKRI). It forms nanospheres around 170 nm, which increase slightly after loading with siRNA and DOX. Notably, it can be internalized by cancer cells mainly through electronic interactions and PD-L1-mediated endocytosis. Compared with poly-l-lysine and polyethyleneimine, it has a much higher efficiency (about four times) of gene transduction while lowering toxicity. In the treatment of cancer, it causes apoptosis (21%) and inhibits the expression of SURVIVIN protein in vitro. In vivo, it shows good biocompatibility as there are no changes in mice's body weight. When administering peptide-siRNA-DOX, tumor growth is inhibited the most (about three times). These results above prove the sequence to be a good candidate for gene therapy and drug delivery.


Subject(s)
B7-H1 Antigen/metabolism , Cell-Penetrating Peptides , Doxorubicin , Drug Delivery Systems , Endocytosis , Nanospheres , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , RNA, Small Interfering , Animals , B7-H1 Antigen/genetics , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanospheres/chemistry , Nanospheres/therapeutic use , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Survivin/genetics , Survivin/metabolism
6.
Adv Mater ; 33(45): e2104779, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34751990

ABSTRACT

Nanoparticle-based small interfering RNA (siRNA) therapy shows great promise for glioblastoma (GBM). However, charge associated toxicity and limited blood-brain-barrier (BBB) penetration remain significant challenges for siRNA delivery for GBM therapy. Herein, novel cation-free siRNA micelles, prepared by the self-assembly of siRNA-disulfide-poly(N-isopropylacrylamide) (siRNA-SS-PNIPAM) diblock copolymers, are prepared. The siRNA micelles not only display enhanced blood circulation time, superior cell take-up, and effective at-site siRNA release, but also achieve potent BBB penetration. Moreover, due to being non-cationic, these siRNA micelles exert no charge-associated toxicity. Notably, these desirable properties of this novel RNA interfering (RNAi) nanomedicine result in outstanding growth inhibition of orthotopic U87MG xenografts without causing adverse effects, achieving remarkably improved survival benefits. Moreover, as a novel type of polymeric micelle, the siRNA micelle displays effective drug loading ability. When utilizing temozolomide (TMZ) as a model loading drug, the siRNA micelle realizes effective synergistic therapy effect via targeting the key gene (signal transducers and activators of transcription 3, STAT3) in TMZ drug resistant pathways. The authors' results show that this siRNA micelle nanoparticle can serve as a robust and versatile drug codelivery platform, and RNAi nanomedicine and for effective GBM treatment.


Subject(s)
Drug Carriers/chemistry , Micelles , Nanomedicine , RNA, Small Interfering/chemistry , Acrylic Resins/chemistry , Animals , Blood-Brain Barrier/metabolism , Carbocyanines/chemistry , Cations/chemistry , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Nanoparticles/chemistry , RNA Interference , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/therapeutic use , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Temozolomide/therapeutic use , Tissue Distribution , Transplantation, Heterologous
7.
Biomed Pharmacother ; 143: 112227, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34563953

ABSTRACT

Primary and secondary prevention protocols aim at reducing the plasma levels of lipids - with particular reference to low-density lipoprotein cholesterol (LDL-C) plasma concentrations - in order to improve the overall survival and reduce the occurrence of major adverse cardiovascular events. The use of statins has been widely considered as the first-line approach in lipids management as they can dramatically impact on the cardiovascular risk profile of individuals. The introduction of ezetimibe and proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors overcame the adverse effects of statins and ameliorate the achievement of the target lipids levels. Indeed, advances in therapies promote the use of specific molecules - i.e. short strands of RNA named small-interfering RNAs (siRNAs) - to suppress the transcription of genes related to lipids metabolism. Recently, the inclisiran has been developed: this is a siRNA able to block the mRNA of the PCSK9 gene. About 50% reduction in low-density lipoprotein cholesterol levels have been observed in randomized controlled trials with inclisiran. The aim of this review was to summarize the literature regarding inclisiran and its possible role in the general management of patients with lipid disorders and/or in primary/secondary prevention protocols.


Subject(s)
Cardiovascular Diseases/prevention & control , Cholesterol, LDL/blood , Dyslipidemias/therapy , Proprotein Convertase 9/metabolism , RNA, Small Interfering/therapeutic use , RNAi Therapeutics , Animals , Biomarkers/blood , Cardiovascular Diseases/epidemiology , Down-Regulation , Dyslipidemias/enzymology , Dyslipidemias/epidemiology , Dyslipidemias/genetics , Heart Disease Risk Factors , Humans , Primary Prevention , Proprotein Convertase 9/genetics , RNA, Small Interfering/adverse effects , RNA, Small Interfering/pharmacokinetics , Risk Assessment , Secondary Prevention , Treatment Outcome
8.
Mol Pharm ; 18(11): 3990-3998, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34591491

ABSTRACT

The study aims to investigate the in vivo distribution, antitumor effect, and safety of cell membrane-penetrating peptide-modified disulfide bond copolymer nanoparticles loaded with small-interfering RNA (siRNA) targeting epidermal growth factor receptor (EGFR) and bromodomain-containing protein 4 (BRD4) in triple-negative breast cancer (TNBC). Polyethylene glycol disulfide bond-linked polyethylenimine (PEG-SS-PEI) was modified with peptides GALA and CREKA and used as vectors to prepare siRNA nanoparticles. The GALA- and CREKA-modified PEG-SS-PEI nanoparticles (GC-NPs) were prepared by mixing siEGFR and siBRD4 (1:1) with GALA-PEG-SS-PEI and CREKA-PEG-SS-PEI (1:1) in an aqueous solution at an N/P ratio of 30:1. Nanoparticles loaded with scrambled siRNA were prepared with the same method. The gene silencing effect on EGFR and BRD4 in vitro was evaluated by Western blotting analysis. TNBC xenograft models were established by subcutaneous injection of MDA-MB-231 cells into female nude mice. At 1, 3, 6, 12, and 24 h after administration of five formulations of Cy5-siRNA (133 µg/10 g) via the tail vein, the mice were observed and imaged for a biodistribution study using an in vivo imaging system. In the pharmacodynamics experiment, tumor-bearing mice were treated with respective siRNA preparations at a dose of 133 µg/10 g for 18 days, and the body weight and tumor size were recorded every other day. The protein expression levels of EGFR, p-EGFR, PI3K, p-PI3K, Akt, p-Akt, BRD4, and c-Myc were determined using Western blotting analysis. Hematological and serum biochemical parameters, organ indices, and HE staining results for the heart, liver, spleen, lung, and kidney were analyzed to evaluate the safety of the nanoparticles. GC-NPs loaded with siEGFR and siBRD4 significantly inhibited the expression of EGFR and BRD4 in vitro. The strongest fluorescence signals were observed in the GC-NP group, especially in tumors, indicating the excellent tumor-targeted delivery of GC-NPs we constructed. Tumor growth was significantly inhibited in the GC-NP-treated group, and the expression of EGFR, p-EGFR, PI3K, p-PI3K, Akt, p-Akt, BRD4, and c-Myc in the tumors decreased by 71%, 68%, 61%, 68%, 48%, 58%, 59%, and 74% compared to the control group, respectively. There was no significant change in hematological parameters, biochemical indices, or tissue morphology in GC-NP-treated mice. SiRNA cotargeting EGFR and BRD4 delivered by GALA- and CREKA-modified PEG-SS-PEI had favorable antitumor effects in vivo toward TNBC with tumor-targeting efficacy and good biocompatibility.


Subject(s)
Genetic Therapy/methods , Nanoparticle Drug Delivery System/chemistry , RNA, Small Interfering/administration & dosage , Triple Negative Breast Neoplasms/therapy , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Gene Silencing , Humans , Mice , Oxidation-Reduction , Polyethylene Glycols/chemistry , Polyethyleneimine/analogs & derivatives , Polyethyleneimine/chemistry , RNA, Small Interfering/pharmacokinetics , Tissue Distribution , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
9.
J Pharmacol Exp Ther ; 379(2): 134-146, 2021 11.
Article in English | MEDLINE | ID: mdl-34413198

ABSTRACT

Conjugation of small interfering RNA (siRNA) to tris N-acetylgalactosamine [(GalNAc)3] can enable highly selective, potent, and durable knockdown of targeted proteins in the liver. However, potential knowledge gaps between in vitro experiments, preclinical species, and clinical scenarios remain. A minimal physiologically based pharmacokinetic-pharmacodynamic model for GalNAc-conjugated siRNA (GalNAc-siRNA) was developed using published data for fitusiran (ALN-AT3), an investigational compound targeting liver antithrombin (AT), to delineate putative determinants governing the whole-body-to-cellular pharmacokinetic (PK) and pharmacodynamic (PD) properties of GalNAc-siRNA and facilitate preclinical-to-clinical translation. The model mathematically linked relevant mechanisms: 1) hepatic biodistribution, 2) tris-GalNAc binding to asialoglycoprotein receptors (ASGPRs) on hepatocytes, 3) ASGPR endocytosis and recycling, 4) endosomal transport and escape of siRNA, 5) cytoplasmic RNA-induced silencing complex (RISC) loading, 6) degradation of target mRNA by bound RISC, and 7) knockdown of protein. Physiologic values for 36 out of 48 model parameters were obtained from the literature. Kinetic parameters governing (GalNAc)3-ASGPR binding and internalization were derived from published studies of uptake in hepatocytes. The proposed model well characterized reported pharmacokinetics, RISC dynamics, and knockdown of AT mRNA and protein by ALN-AT3 in mice. The model bridged multiple PK-PD data sets in preclinical species (mice, rat, monkey) and successfully captured reported plasma pharmacokinetics and AT knockdown in a phase I ascending-dose study. Estimates of in vivo potency were similar (∼2-fold) across species. Subcutaneous absorption and serum AT degradation rate constants scaled across species by body weight with allometric exponents of -0.29 and -0.22. The proposed mechanistic modeling framework characterizes the unique PK-PD properties of GalNAc-siRNA. SIGNIFICANCE STATEMENT: Tris N-acetylgalactosamine (GalNAc)3-conjugated small interfering RNA (siRNA) therapeutics enable liver-targeted gene therapy and precision medicine. Using a translational and systems-based minimal physiologically based pharmacokinetic-pharmacodynamic (mPBPK-PD) modeling approach, putative determinants influencing GalNAc-conjugated siRNA (GalNAc-siRNA) functionality in three preclinical species and humans were investigated. The developed model successfully integrated and characterized relevant published in vitro-derived biomeasures, mechanistic PK-PD profiles in animals, and observed clinical PK-PD responses for an investigational GalNAc-siRNA (fitusiran). This modeling effort delineates the disposition and liver-targeted pharmacodynamics of GalNAc-siRNA.


Subject(s)
Acetylgalactosamine/pharmacokinetics , Gene Silencing/physiology , Models, Biological , RNA, Small Interfering/pharmacokinetics , Acetylgalactosamine/genetics , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Haplorhini , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Mice , RNA, Small Interfering/genetics , Rats , Tissue Distribution/drug effects , Tissue Distribution/physiology
10.
Nat Commun ; 12(1): 3953, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172725

ABSTRACT

The systemic therapeutic utilisation of RNA interference (RNAi) is limited by the non-specific off-target effects, which can have severe adverse impacts in clinical applications. The accurate use of RNAi requires tumour-specific on-demand conditional activation to eliminate the off-target effects of RNAi, for which conventional RNAi systems cannot be used. Herein, a tumourous biomarker-activated RNAi platform is achieved through the careful design of RNAi prodrugs in extracellular vesicles (EVs) with cancer-specific recognition/activation features. These RNAi prodrugs are assembled by splitting and reconstituting the principal siRNAs into a hybridisation chain reaction (HCR) amplification machine. EVs facilitate the specific and efficient internalisation of RNAi prodrugs into target tumour cells, where endogenous microRNAs (miRNAs) promote immediate and autonomous HCR-amplified RNAi activation to simultaneously silence multiantenna hypoxia-related genes. With multiple guaranteed cancer recognition and synergistic therapy features, the miRNA-initiated HCR-promoted RNAi cascade holds great promise for personalised theranostics that enable reliable diagnosis and programmable on-demand therapy.


Subject(s)
Hypoxia/genetics , Precision Medicine , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/therapeutic use , Extracellular Vesicles/chemistry , Extracellular Vesicles/transplantation , Gene Silencing , Humans , Mice , MicroRNAs/genetics , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics
11.
Clin J Am Soc Nephrol ; 16(7): 1025-1036, 2021 07.
Article in English | MEDLINE | ID: mdl-33985991

ABSTRACT

BACKGROUND AND OBJECTIVES: In the rare disease primary hyperoxaluria type 1, overproduction of oxalate by the liver causes kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. Lumasiran, an RNA interference therapeutic, suppresses glycolate oxidase, reducing hepatic oxalate production. The objective of this first-in-human, randomized, placebo-controlled trial was to evaluate the safety, pharmacokinetic, and pharmacodynamic profiles of lumasiran in healthy participants and patients with primary hyperoxaluria type 1. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This phase 1/2 study was conducted in two parts. In part A, healthy adults randomized 3:1 received a single subcutaneous dose of lumasiran or placebo in ascending dose groups (0.3-6 mg/kg). In part B, patients with primary hyperoxaluria type 1 randomized 3:1 received up to three doses of lumasiran or placebo in cohorts of 1 or 3 mg/kg monthly or 3 mg/kg quarterly. Patients initially assigned to placebo crossed over to lumasiran on day 85. The primary outcome was incidence of adverse events. Secondary outcomes included pharmacokinetic and pharmacodynamic parameters, including measures of oxalate in patients with primary hyperoxaluria type 1. Data were analyzed using descriptive statistics. RESULTS: Thirty-two healthy participants and 20 adult and pediatric patients with primary hyperoxaluria type 1 were enrolled. Lumasiran had an acceptable safety profile, with no serious adverse events or study discontinuations attributed to treatment. In part A, increases in mean plasma glycolate concentration, a measure of target engagement, were observed in healthy participants. In part B, patients with primary hyperoxaluria type 1 had a mean maximal reduction from baseline of 75% across dosing cohorts in 24-hour urinary oxalate excretion. All patients achieved urinary oxalate levels ≤1.5 times the upper limit of normal. CONCLUSIONS: Lumasiran had an acceptable safety profile and reduced urinary oxalate excretion in all patients with primary hyperoxaluria type 1 to near-normal levels. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER: Study of Lumasiran in Healthy Adults and Patients with Primary Hyperoxaluria Type 1, NCT02706886.


Subject(s)
Hyperoxaluria, Primary/drug therapy , Oxalates/urine , RNA, Small Interfering/pharmacology , RNA, Small Interfering/pharmacokinetics , Renal Agents/pharmacology , Renal Agents/pharmacokinetics , Adolescent , Adult , Child , Female , Glycolates/blood , Humans , Hyperoxaluria, Primary/blood , Hyperoxaluria, Primary/urine , Male , RNA, Small Interfering/adverse effects , Renal Agents/adverse effects , Single-Blind Method , Young Adult
12.
Nat Commun ; 12(1): 2928, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006888

ABSTRACT

Small interfering RNA (siRNA) is an effective therapeutic to regulate the expression of target genes in vitro and in vivo. Constructing a siRNA delivery system with high serum stability, especially responsive to endogenous stimuli, remains technically challenging. Herein we develop anti-degradation Y-shaped backbone-rigidified triangular DNA bricks with sticky ends (sticky-YTDBs) and tile them onto a siRNA-packaged gold nanoparticle in a programmed fashion, forming a multi-functional three-dimensional (3D) DNA shell. After aptamers are arranged on the exterior surface, a biocompatible siRNA-encapsulated core/shell nanoparticle, siRNA/Ap-CS, is achieved. SiRNAs are internally encapsulated in a 3D DNA shell and are thus protected from enzymatic degradation by the outermost layer of YTDB. The siRNAs can be released by endogenous miRNA and execute gene silencing within tumor cells, causing cell apoptosis higher than Lipo3000/siRNA formulation. In vivo treatment shows that tumor growth is completely (100%) inhibited, demonstrating unique opportunities for next-generation anticancer-drug carriers for targeted cancer therapies.


Subject(s)
DNA/chemistry , Gene Transfer Techniques , Gold/chemistry , Metal Nanoparticles/chemistry , Neoplasms/genetics , RNA, Small Interfering/genetics , A549 Cells , Animals , DNA/genetics , Gene Silencing , HeLa Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplasms/metabolism , Neoplasms/therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , Xenograft Model Antitumor Assays/methods
13.
ACS Appl Mater Interfaces ; 13(20): 23396-23409, 2021 May 26.
Article in English | MEDLINE | ID: mdl-33982563

ABSTRACT

Paclitaxel (PTX) is a first-line chemotherapeutic drug for breast cancer, but PTX resistance often occurs in metastatic breast cancer. In addition, due to the poor targeting of chemotherapeutic drugs and the presence of the blood-brain barrier (BBB), it is hard to effectively treat brain metastatic breast cancer using paclitaxel. Thus, it is urgent to develop an effective drug delivery system for the treatment of brain metastatic breast cancer. The current study found that TWF1 gene, an epithelial-mesenchymal transition-associated gene, was overexpressed in brain metastatic breast cancer (231-BR) cells and was associated with the PTX resistance of 231-BR cells. Knockdown of TWF1 by small interference RNA (siRNA) in 231-BR cells could effectively increase the sensitivity of brain metastatic breast cancer cells to paclitaxel. Then, a liposome-based drug delivery system was developed for PTX delivery across BBB, enhancing PTX sensitivity and brain metastases targeting via BRBP1 peptide modification. The results showed that BRBP1-modified liposomes could effectively cross the BBB, specifically accumulate in brain metastases, and effectively interfere TWF1 gene expression in vitro and in vivo, and thus they enhanced proliferation inhibition, cell cycle arrest, and apoptosis induction, thereby inhibiting the formation and growth of brain metastases. In summary, our results indicated that BRBP1-modified and PTX- and TWF1 siRNA-loaded liposomes have the potential for the treatment of brain metastatic breast cancer, which lays the foundation for the development of a new targeted drug delivery system.


Subject(s)
Brain Neoplasms , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Liposomes , Paclitaxel , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Cell Line, Tumor , Female , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Mice , Mice, Inbred BALB C , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , Oligopeptides/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
14.
Molecules ; 26(6)2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33808586

ABSTRACT

Here, a novel strategy of formulating efficient polymeric carriers based on the already described INU-IMI-DETA for gene material whose structural, functional, and biological properties can be modulated and improved was successfully investigated. In particular, two novel derivatives of INU-IMI-DETA graft copolymer were synthesized by chemical functionalisation with epidermal growth factor (EGF) or polyethylenglycol (PEG), named INU-IMI-DETA-EGF and INU-IMI-DETA-PEG, respectively, in order to improve the performance of already described "inulin complex nanoaggregates" (ICONs). The latter were thus prepared by appropriately mixing the two copolymers, by varying each component from 0 to 100 wt% on the total mixture, named EP-ICONs. It was seen that the ability of the INU-IMI-DETA-EGF/INU-IMI-DETA-PEG polymeric mixture to complex siGL3 increases with the increase in the EGF-based component in the EP-ICONs and, for each sample, with the increase in the copolymer:siRNA weight ratio (R). On the other hand, the susceptibility of loaded siRNA towards RNase decreases with the increase in the pegylated component in the polymeric mixture. At all R values, the average size and the zeta potential values are suitable for escaping from the RES system and suitable for prolonged intravenous circulation. By means of biological characterisation, it was shown that MCF-7 cells are able to internalize mainly the siRNA-loaded into EGF-decorated complexes, with a significant difference from ICONs, confirming its targeting function. The targeting effect of EGF on EP-ICONs was further demonstrated by a competitive cell uptake study, i.e., after cell pre-treatment with EGF. Finally, it was shown that the complexes containing both EGF and PEG are capable of promoting the internalisation and therefore the transfection of siSUR, a siRNA acting against surviving mRNA, and to increase the sensitivity to an anticancer agent, such as doxorubicin.


Subject(s)
Drug Carriers , Inulin , Nanostructures , Neoplasms/diet therapy , RNA, Small Interfering , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Inulin/chemistry , Inulin/pharmacokinetics , Inulin/pharmacology , MCF-7 Cells , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoplasms/metabolism , Neoplasms/pathology , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
15.
Mol Pharm ; 18(4): 1491-1506, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33734715

ABSTRACT

RNA-based therapeutics have emerged as one of the most powerful therapeutic options used for the modulation of gene/protein expression and gene editing with the potential to treat neurodegenerative diseases. However, the delivery of nucleic acids to the central nervous system (CNS), in particular by the systemic route, remains a major hurdle. This review will focus on the strategies for systemic delivery of therapeutic nucleic acids designed to overcome these barriers. Pathways and mechanisms of transport across the blood-brain barrier which could be exploited for delivery are described, focusing in particular on smaller nucleic acids including antisense oligonucleotides (ASOs) and small interfering RNA (siRNA). Approaches used to enhance delivery including chemical modifications, nanocarrier systems, and target selection (cell-specific delivery) are critically analyzed. Learnings achieved from a comparison of the successes and failures reported for CNS delivery of ASOs versus siRNA will help identify opportunities for a wider range of nucleic acids and accelerate the clinical translation of these innovative therapies.


Subject(s)
Central Nervous System Diseases/therapy , Drug Carriers/chemistry , Genetic Therapy/methods , Oligonucleotides, Antisense/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Blood-Brain Barrier/metabolism , Central Nervous System Diseases/genetics , Disease Models, Animal , Drug Compounding , Humans , Nanoparticles/chemistry , Oligonucleotides, Antisense/pharmacokinetics , Permeability , RNA, Small Interfering/pharmacokinetics
16.
Endocrinology ; 162(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33411885

ABSTRACT

Extracellular vesicles (EVs) are key players of intercellular communication in the physiological and pathological setting. In cancer, EVs mediate complex signaling mechanisms between cancer cells and the tumor microenvironment (TME), and can influence tumor progression and the response to existing therapies. Importantly, EVs can be loaded with therapeutic agents and modified to display tumor-targeting molecules. In the field of nanomedicine, EVs have been engineered to serve as therapeutic delivery vehicles for several anticancer agents, including antibodies, chemotherapy, compounds, CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats-associated endonuclease 9), and small interfering RNA (siRNA). Notably, the engineered EVs were shown to suppress malignant features of cancer cells, to elicit antitumor immunity, and to decrease tumor angiogenesis. Here, we review the EV-based therapies designed to target cancer cells and to educate components of the TME to drive antitumor responses. These studies illustrate the multifunctional applications of EVs in the development of anticancer therapies and their translational potential for cancer treatment.


Subject(s)
Drug Delivery Systems , Extracellular Vesicles/physiology , Neoplasms/therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Bioengineering/methods , Bioengineering/trends , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Neoplasms/metabolism , Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , Tumor Microenvironment/drug effects
17.
Mol Pharm ; 18(2): 641-666, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32453574

ABSTRACT

Diabetic nephropathy (DN), a chronic progressive kidney disease, is a significant complication of diabetes mellitus. Dysregulation of the histone deacetylases (HDACs) gene has been implicated in the pathogenesis of DN. Hence, the HDAC-inhibitors have emerged as a critical class of therapeutic agents in DN; however, the currently available HDAC4-inhibitors are mostly nonselective in nature as well as inhibit multiple HDACs. RNA interference of HDAC4 (HDAC4 siRNA) has shown immense promise, but the clinical translation has been impeded due to lack of a targeted, specific, and in vivo applicable delivery modality. In the present investigation, we examined Cyclo(RGDfC) (cRGD) truncated polymeric nanoplex with dendrimeric templates for targeted HDAC4 Gene Silencing. The developed nanoplex exhibited enhanced encapsulation of siRNA and offered superior protection against serum RNase nucleases degradation. The nanoplex was tested on podocytes (in vitro), wherein it showed selective binding to the αvß3 integrin receptor, active cellular uptake, and significant in vitro gene silencing. The in vivo experiments showed remarkable suppression of the HDAC4 and inhibition in the progression of renal fibrosis in the Streptozotocin (STZ) induced DN C57BL/6 mice model. Histopathological and toxicological studies revealed nonsignificant abnormality/toxicity with the nanoplex. Conclusively, nanoplex was found as a promising tactic for targeted therapy of podocytes and could be extended for other kidney-related ailments.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Drug Carriers/chemistry , Histone Deacetylase Inhibitors/administration & dosage , Oligopeptides/chemistry , Animals , Dendrimers/chemistry , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Drug Stability , Gene Silencing , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Kidney/drug effects , Kidney/pathology , Male , Mice , Nanoparticles/chemistry , Peptides, Cyclic/chemistry , Podocytes , Primary Cell Culture , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects , Streptozocin/administration & dosage , Streptozocin/toxicity
18.
Eur J Pharm Biopharm ; 158: 284-293, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33285246

ABSTRACT

The delivery of oligonucleotides such as siRNA to the lung is a major challenge, as this group of drugs has difficulties to overcome biological barriers due to its polyanionic character and the associated hydrophilic properties, resulting in inefficient delivery. Especially in diseases such as asthma, chronic obstructive pulmonary disease and cystic fibrosis, where increased proinflammation is present, a targeted RNA therapy is desirable due to the high potency of these oligonucleotides. To address these problems and to ensure efficient uptake of siRNA in macrophages, a microparticulate, cylindrical delivery system was developed. In the first step, this particle system was tested for its aerodynamic characteristics to evaluate the aerodynamic properties to optimize lung deposition. The mass median aerodynamic diameter of 2.52 ± 0.23 µm, indicates that the desired target should be reached. The inhibition of TNF-α release, as one of the main mediators of proinflammatory reactions, was investigated. We could show that our carrier system can be loaded with siRNA against TNF-α. Gel electrophoreses allowed to demonstrate that the load can be incorporated and released without being degraded. The delivery system was found to transport a mass fraction of 0.371% [%w/w] as determined by inductively coupled plasma mass spectroscopy. When investigating the release kinetics, the results showed that several days are necessary to release a major amount of the siRNA indicating a sustained release. The cylindrical microparticles with an aspect ratio of 3.3 (ratio of length divided by width) were then tested in vitro successfully reducing TNF-α release from human macrophages significantly by more than 30%. The developed formulation presents a possible oligonucleotide delivery system allowing due to its internal structure to load and protect siRNA.


Subject(s)
Drug Carriers/chemistry , Macrophages/metabolism , RNA, Small Interfering/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Administration, Inhalation , Asthma/drug therapy , Cystic Fibrosis/drug therapy , Drug Compounding/methods , Drug Liberation , Humans , Microspheres , Molecular Targeted Therapy/methods , Particle Size , Pulmonary Disease, Chronic Obstructive/drug therapy , RNA, Small Interfering/pharmacokinetics , THP-1 Cells
19.
Mol Med Rep ; 22(5): 4183-4196, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33000194

ABSTRACT

Polyethylene glycol (PEG)­modifications (PEGylations) of cationic liposome/small interfering RNA complexes (siRNA lipoplexes) can enhance their systemic stability. The present study determined the effects of PEG anchors in PEGylated siRNA lipoplexes on in vitro gene­silencing effects and siRNA biodistribution after intravenous injection. Three types of dialkyl or trialkyl cationic lipids were used in the current study for the preparation of cationic liposomes. Additionally, various PEGylated siRNA lipoplexes that contained PEG­1,2­distearoyl­sn­-glycero­-3­phosphoethanolamine (DSPE), PEG­1,2­distearoyl­rac­glycero­3­-methylpolyoxyethylene (DSG), PEG­cholesterol (PEG­Chol) and PEG­chondroitin sulfate conjugate (PEG­CS) were prepared. The results revealed that PEGylation of siRNA lipoplexes with PEG­DSPE strongly decreased gene­silencing effects in cells. In contrast, those with PEG­DSG, PEG­Chol and PEG­CS did not largely decrease gene-silencing effects. However, regardless of the PEG­derivative type, PEGylation of siRNA lipoplexes decreased their agglutination with erythrocytes. Furthermore, intravenous injection of PEGylated siRNA lipoplexes markedly decreased the accumulation of siRNA in the lungs, regardless of the type of PEG­derivative. However, non­PEGylated siRNA lipoplexes accumulated mainly in the lungs regardless of the siRNA lipoplex cationic lipid type. The results indicated that PEGylation of siRNA lipoplexes with PEG­DSG, PEG­Chol and PEG­CS may improve systemic stability without losing transfection activity by PEGylation.


Subject(s)
Lung/chemistry , Polyethylene Glycols/chemistry , RNA, Small Interfering/pharmacokinetics , Administration, Intravenous , Animals , Cholesterol/analogs & derivatives , Cholesterol/chemistry , Chondroitin Sulfates/chemistry , Female , Gene Silencing , Humans , Liposomes , MCF-7 Cells , Mice , Phosphatidylethanolamines/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Tissue Distribution
20.
Methods Mol Biol ; 2176: 141-154, 2020.
Article in English | MEDLINE | ID: mdl-32865788

ABSTRACT

Oligonucleotide drugs (ODs) have gained increasing attention owing to their promising therapeutic potential. One major obstacle that ODs have been facing is the lack of appropriate in vitro validation systems that can predict in vivo activity and toxicity. We have devised a transfection method called CEM (Ca2+-enrichment method), where the simple enrichment of calcium ion with calcium chloride in culture medium potentiates the activity of various types of naked oligonucleotides including gapmers, siRNA, and phosphorodiamidate morpholino antisense oligonucleotides (PMO) in many cultured cell lines with limited cytotoxicity. We here describe a precise procedure of the method. Besides the benefit of the CEM's predictive power to accurately estimate in vivo activity of ODs of your interest in drug discovery and development settings, this cost-efficient, easy-to-access method can be a robust laboratory technique to modulate gene expressions with ODs with a variety of mechanisms of action.


Subject(s)
Calcium/pharmacology , Cell Membrane Permeability/drug effects , Oligonucleotides/chemistry , Oligonucleotides/genetics , Transfection/methods , A549 Cells , Animals , Base Sequence/physiology , Cell Culture Techniques , Cell Line , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Morpholinos/chemistry , Morpholinos/genetics , Morpholinos/pharmacokinetics , Nucleic Acid Conformation/drug effects , Oligonucleotides/pharmacokinetics , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacokinetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Reverse Transcriptase Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...