Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.704
Filtrer
1.
Nihon Yakurigaku Zasshi ; 159(4): 254-263, 2024.
Article de Japonais | MEDLINE | ID: mdl-38945909

RÉSUMÉ

Inclisiran sodium (Brand name: LEQVIO® for s.c. injection syringe 300 |mg, hereinafter referred to as inclisiran), a small interfering ribonucleic acid (siRNA) product that targets the mRNA that encodes the proprotein convertase subtilisin/kexin type 9 (PCSK9) protein was approved on September 25, 2023 for the indication of "Familial hypercholesterolemia, hypercholesterolemia" in Japan. Inclisiran is conjugated on the sense strand with triantennary N-acetylgalactosamine to facilitate uptake by hepatocytes. In vitro and in vivo pharmacology studies demonstrated the lowering effects of PCSK9 and LDL-C in hepatocytes and cynomolgus monkeys. It was considered unlikely to cause clinically significant risks due to toxicities arising from complementary binding to non-target RNA sequences (hybridization-dependent off-target effects). Clinical trials conducted globally including Japan in patients with familial hypercholesterolemia and hypercholesterolemia who did not reach the LDL-C target showed that inclisiran sodium 300 |mg dosed at Day 1, Day 90 and then every 6 months demonstrated significant LDL-C reduction and the efficacy sustained long. The majority of patients achieved the guideline recommended LDL-C targets. Inclisiran sodium 300 |mg was well tolerated and there were no specific safety concerns. Therefore, inclisiran is expected to be a new therapeutic option for the patients with familial hypercholesterolemia and hypercholesterolemia.


Sujet(s)
Cholestérol LDL , Proprotéine convertase 9 , Petit ARN interférent , Humains , Cholestérol LDL/sang , Animaux , Petit ARN interférent/administration et posologie , Petit ARN interférent/pharmacologie , Proprotéine convertase 9/métabolisme , Proprotéine convertase 9/génétique , Hypercholestérolémie/traitement médicamenteux , Hyperlipoprotéinémie de type II/traitement médicamenteux , Hyperlipoprotéinémie de type II/génétique
2.
Signal Transduct Target Ther ; 9(1): 150, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38902241

RÉSUMÉ

This study aimed to develop a pan-genotypic and multifunctional small interfering RNA (siRNA) against hepatitis B virus (HBV) with an efficient delivery system for treating chronic hepatitis B (CHB), and explore combined RNA interference (RNAi) and immune modulatory modalities for better viral control. Twenty synthetic siRNAs targeting consensus motifs distributed across the whole HBV genome were designed and evaluated. The lipid nanoparticle (LNP) formulation was optimized by adopting HO-PEG2000-DMG lipid and modifying the molar ratio of traditional polyethylene glycol (PEG) lipid in LNP prescriptions. The efficacy and safety of this formulation in delivering siHBV (tLNP/siHBV) along with the mouse IL-2 (mIL-2) mRNA (tLNP/siHBVIL2) were evaluated in the rAAV-HBV1.3 mouse model. A siRNA combination (terms "siHBV") with a genotypic coverage of 98.55% was selected, chemically modified, and encapsulated within an optimized LNP (tLNP) of high efficacy and security to fabricate a therapeutic formulation for CHB. The results revealed that tLNP/siHBV significantly reduced the expression of viral antigens and DNA (up to 3log10 reduction; vs PBS) in dose- and time-dependent manners at single-dose or multi-dose frequencies, with satisfactory safety profiles. Further studies showed that tLNP/siHBVIL2 enables additive antigenic and immune control of the virus, via introducing potent HBsAg clearance through RNAi and triggering strong HBV-specific CD4+ and CD8+ T cell responses by expressed mIL-2 protein. By adopting tLNP as nucleic acid nanocarriers, the co-delivery of siHBV and mIL-2 mRNA enables synergistic antigenic and immune control of HBV, thus offering a promising translational therapeutic strategy for treating CHB.


Sujet(s)
Virus de l'hépatite B , Interleukine-2 , Nanoparticules , Petit ARN interférent , Animaux , Souris , Virus de l'hépatite B/génétique , Interleukine-2/génétique , Interleukine-2/immunologie , Interleukine-2/pharmacologie , Humains , Petit ARN interférent/génétique , Petit ARN interférent/administration et posologie , Nanoparticules/composition chimique , ARN messager/génétique , Hépatite B chronique/thérapie , Hépatite B chronique/génétique , Hépatite B chronique/virologie , Interférence par ARN , Hépatite B/thérapie , Hépatite B/génétique , Hépatite B/virologie , Thérapie par l'interférence par ARN , Liposomes
3.
Methods Mol Biol ; 2822: 353-365, 2024.
Article de Anglais | MEDLINE | ID: mdl-38907928

RÉSUMÉ

Polymeric delivery systems could enable the fast- and low-side-effect transport of various RNA classes. Previously, we demonstrated that polyvinylamine (PVAm), a cationic polymer, transfects many kinds of RNAs with high efficiency and low toxicity both in vitro and in vivo. The modification of poly lactic-co-glycolic acid (PLGA) with cartilage-targeting peptide (CAP) enhances its stiffness and tissue-specific delivery of RNA to overcome the avascular nature of articular cartilage. Here we describe the protocol to use PVAm as an RNA carrier, and further, by modifying PVAm with PLGA and CAP, the corresponding co-polymer could be applied for functional RNA delivery for osteoarthritis treatment.


Sujet(s)
Copolymère d'acide poly(lactique-co-glycolique) , Polyvinyles , Polyvinyles/composition chimique , Animaux , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Humains , Acide lactique/composition chimique , Transfection/méthodes , Techniques de transfert de gènes , Acide polyglycolique/composition chimique , Vecteurs de médicaments/composition chimique , Petit ARN interférent/administration et posologie , Petit ARN interférent/génétique , Arthrose/traitement médicamenteux
4.
Mol Biol Rep ; 51(1): 737, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874790

RÉSUMÉ

Acute lymphoblastic leukemia (ALL) is the most common type of cancer among children, presenting significant healthcare challenges for some patients, including drug resistance and the need for targeted therapies. SiRNA-based therapy is one potential solution, but problems can arise in administration and the need for a delivery system to protect siRNA during intravenous injection. Additionally, siRNA encounters instability and degradation in the reticuloendothelial system, off-target effects, and potential immune system stimulation. Despite these limitations, some promising results about siRNA therapy in ALL patients have been published in recent years, showing the potential for more effective and precise treatment, reduced side effects, and personalized approaches. While siRNA-based therapies demonstrate safety and efficacy, addressing the mentioned limitations is crucial for further optimization. Advancements in siRNA-delivery technologies and combination therapies hold promise to improve treatment effectiveness and overcome drug resistance. Ultimately, despite its challenges, siRNA therapy has the potential to revolutionize ALL treatments and improve patient outcomes.


Sujet(s)
Leucémie-lymphome lymphoblastique à précurseurs B et T , Petit ARN interférent , Humains , Petit ARN interférent/génétique , Petit ARN interférent/administration et posologie , Petit ARN interférent/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Thérapie génétique/méthodes , Animaux , Résistance aux médicaments antinéoplasiques/génétique
5.
Nat Commun ; 15(1): 5398, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926348

RÉSUMÉ

Ionotropic gelation is widely used to fabricate targeting nanoparticles (NPs) with polysaccharides, leveraging their recognition by specific lectins. Despite the fabrication scheme simply involves self-assembly of differently charged components in a straightforward manner, the identification of a potent combinatory formulation is usually limited by structural diversity in compound collections and trivial screen process, imposing crucial challenges for efficient formulation design and optimization. Herein, we report a diversity-oriented combinatory formulation screen scheme to identify potent gene delivery cargo in the context of precision cardiac therapy. Distinct categories of cationic compounds are tested to construct RNA delivery system with an ionic polysaccharide framework, utilizing a high-throughput microfluidics workstation coupled with streamlined NPs characterization system in an automatic, step-wise manner. Sequential computational aided interpretation provides insights in formulation optimization in a broader scenario, highlighting the usefulness of compound library diversity. As a result, the out-of-bag NPs, termed as GluCARDIA NPs, are utilized for loading therapeutic RNA to ameliorate cardiac reperfusion damages and promote the long-term prognosis. Overall, this work presents a generalizable formulation design strategy for polysaccharides, offering design principles for combinatory formulation screen and insights for efficient formulation identification and optimization.


Sujet(s)
Nanoparticules , Polyosides , Polyosides/composition chimique , Nanoparticules/composition chimique , Animaux , Humains , Souris , Techniques de transfert de gènes , Thérapie par l'interférence par ARN/méthodes , Interférence par ARN , Mâle , Petit ARN interférent/administration et posologie , Petit ARN interférent/composition chimique , Souris de lignée C57BL , Lésion de reperfusion myocardique/thérapie
6.
Biomacromolecules ; 25(7): 4177-4191, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38866384

RÉSUMÉ

After RNAi was first discovered over 20 years ago, siRNA-based therapeutics are finally becoming reality. However, the delivery of siRNA has remained a challenge. In our previous research, we found that spermine-based poly(ß-amino ester)s are very promising for siRNA delivery. However, the role of hydrophobic modification in siRNA delivery of spermine-based poly(ß-amino ester)s is not fully understood yet. In the current work, we synthesized spermine-based poly(ß-amino ester)s with different percentages of oleylamine side chains, named P(SpOABAE). The chemical structures of the polymers were characterized by 1H NMR. The polymers showed efficient siRNA encapsulation determined by SYBR Gold assays. The hydrodynamic diameters of the P(SpOABAE) polyplexes from charge ratio N/P 1 to 20 were 30-100 nm except for aggregation phenomena observed at N/P 3. Morphology of the polyplexes was visualized by atomic force microscopy, and cellular uptake was determined by flow cytometry in H1299 cells, where all the polyplexes showed significantly higher cellular uptake than hyperbranched polyethylenimine (25 kDa). The most hydrophobic P(SpOABAE) polyplexes were able to achieve more than 90% GFP knockdown in H1299/eGFP cells. The fact that gene silencing efficacy increased with hydrophobicity but cellular uptake was affected by both charge and hydrophobic interactions highlights the importance of endosomal escape. For pulmonary administration and improved storage stability, the polyplexes were spray-dried. Results confirmed the maintained siRNA activity after storage for 3 months at room temperature, indicating potential for dry powder inhalation.


Sujet(s)
Interactions hydrophobes et hydrophiles , Petit ARN interférent , Spermine , Petit ARN interférent/composition chimique , Petit ARN interférent/administration et posologie , Petit ARN interférent/pharmacologie , Spermine/composition chimique , Humains , Administration par inhalation , Polymères/composition chimique , Poudres/composition chimique , Lignée cellulaire tumorale
7.
Theranostics ; 14(7): 2777-2793, 2024.
Article de Anglais | MEDLINE | ID: mdl-38773978

RÉSUMÉ

Small extracellular vesicles (sEVs) are naturally occurring vesicles that have the potential to be manipulated to become promising drug delivery vehicles for on-demand in vitro and in vivo gene editing. Here, we developed the modular safeEXO platform, a prototype sEV delivery vehicle that is mostly devoid of endogenous RNA and can efficaciously deliver RNA and ribonucleoprotein (RNP) complexes to their intended intracellular targets manifested by downstream biologic activity. We also successfully engineered producer cells to produce safeEXO vehicles that contain endogenous Cas9 (safeEXO-CAS) to effectively deliver efficient ribonucleoprotein (RNP)-mediated CRISPR genome editing machinery to organs or diseased cells in vitro and in vivo. We confirmed that safeEXO-CAS sEVs could co-deliver ssDNA, sgRNA and siRNA, and efficaciously mediate gene insertion in a dose-dependent manner. We demonstrated the potential to target safeEXO-CAS sEVs by engineering sEVs to express a tissue-specific moiety, integrin alpha-6 (safeEXO-CAS-ITGA6), which increased their uptake to lung epithelial cells in vitro and in vivo. We tested the ability of safeEXO-CAS-ITGA6 loaded with EMX1 sgRNAs to induce lung-targeted editing in mice, which demonstrated significant gene editing in the lungs with no signs of morbidity or detectable changes in immune cell populations. Our results demonstrate that our modular safeEXO platform represents a targetable, safe, and efficacious vehicle to deliver nucleic acid-based therapeutics that successfully reach their intracellular targets. Furthermore, safeEXO producer cells can be genetically manipulated to produce safeEXO vehicles containing CRISPR machinery for more efficient RNP-mediated genome editing. This platform has the potential to improve current therapies and increase the landscape of treatment for various human diseases using RNAi and CRISPR approaches.


Sujet(s)
Systèmes CRISPR-Cas , Vésicules extracellulaires , Édition de gène , Techniques de transfert de gènes , Édition de gène/méthodes , Vésicules extracellulaires/métabolisme , Systèmes CRISPR-Cas/génétique , Animaux , Humains , Souris , Ribonucléoprotéines/génétique , Ribonucléoprotéines/métabolisme , Petit ARN interférent/administration et posologie , Petit ARN interférent/génétique , /génétique
8.
Int J Pharm ; 659: 124261, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38782155

RÉSUMÉ

The progression of renal fibrosis to end-stage renal disease (ESRD) is significantly influenced by transforming growth factor-beta (TGF-beta) signal pathway. This study aimed to develop nanoparticles (PMVs@PLGA complexes) with platelet membrane camouflage, which can transport interfering RNA to target and regulate the TGF-ß1 pathway in damaged renal tissues. The aim is to reduce the severity of acute kidney injury and to reduce fibrosis in chronic kidney disease. Hence, we formulated PMVs@TGF-ß1-siRNA NP complexes and employed them for both in vitro and in vivo therapy. From the experimental findings we know that the PMVs@siRNA NPs could effectively target the kidneys in unilateral ureteral obstruction (UUO) mice and ischemia/reperfusion injury (I/R) mice. In animal models of treatment, PMVs@siRNA NP complexes effectively decreased the expression of TGF-ß1 and mitigated inflammation and fibrosis in the kidneys by blocking the TGF-ß1/Smad3 pathway. Therefore, these PMVs@siRNA NP complexes can serve as a promising biological delivery system for treating kidney diseases.


Sujet(s)
Fibrose , Nanoparticules , Petit ARN interférent , Facteur de croissance transformant bêta-1 , Animaux , Petit ARN interférent/administration et posologie , Facteur de croissance transformant bêta-1/métabolisme , Mâle , Souris , Plaquettes/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/prévention et contrôle , Rein/métabolisme , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Obstruction urétérale/thérapie , Matériaux biomimétiques/administration et posologie , Matériaux biomimétiques/composition chimique , Inflammation/traitement médicamenteux , Modèles animaux de maladie humaine , Humains , Protéine Smad-3/métabolisme , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Atteinte rénale aigüe/prévention et contrôle
10.
Nucleic Acids Res ; 52(10): 5423-5437, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38742636

RÉSUMÉ

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.


Sujet(s)
Acétyl-galactosamine , Petit ARN interférent , Animaux , Acétyl-galactosamine/composition chimique , Acétyl-galactosamine/métabolisme , Petit ARN interférent/administration et posologie , Petit ARN interférent/pharmacocinétique , Petit ARN interférent/composition chimique , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Administration par voie orale , Souris , Rats , Interférence par ARN , Mâle , Biodisponibilité , Humains , Rat Sprague-Dawley , Macaca fascicularis , Foie/métabolisme , Macaca mulatta
11.
J Control Release ; 370: 747-762, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38740094

RÉSUMÉ

Rheumatoid arthritis (RA) is a progressive autoimmune disease and drug therapy has been restricted due to poor therapeutic efficacy and adverse effects. In RA synovium, dendritic cells present self-antigens to activate cascade immune pathway. Furthermore, downstream macrophages secrete high levels of pro-inflammatory cytokines; Hyperplasia of activated synovial fibroblasts (FLS) is responsible for hypoxic synovium microenvironment, secretion of cytokines/chemokines and erosion of bone/cartilage tissues. Positive feedback loop of inflammation between macrophages and FLS independent of antigen-presentation is constructed. Herein, an injectable pH-sensitive peptide hydrogel encapsulating siRNA/Methotrexate-polyethyleneimine (siMP, including sip65MP, sip38MP, siCD86MP) and Bismuthene nanosheet/Methotrexate-polyethyleneimine (BiMP) is successfully developed. Among them, siCD86MP reduces protein level of co-stimulatory molecule CD86 while sip65MP and sip38MP separately inhibit NF-κB and MAPK-p38 pathways of macrophages and FLS to suppress secretion of cytokines and MMPs. Meanwhile, reduction in anti-apoptotic property of FLS induced by inhibition of NF-κB pathway has a synergistic effect with photodynamic therapy (PDT) and photothermal therapy (PTT) mediated by BiMP for FLS elimination, effectively ameliorating hypoxic synovium microenvironment. After being injected into synovium, hydrogel responds to acidic microenvironment and serves as a reservoir for sustained drug release and inherent retention capacity of which enables cationic nanoparticles to bypass tissue barrier for precise synovium targeting. This brand-new drug delivery system combines modulating cascade immune pathway from beginning to end by RNAi and eliminating FLS for improving synovium microenvironment by phototherapy together, providing a robust strategy for clinical RA treatment.


Sujet(s)
Polyarthrite rhumatoïde , Fibroblastes , Hydrogels , Méthotrexate , Membrane synoviale , Fibroblastes/effets des médicaments et des substances chimiques , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/thérapie , Hydrogels/administration et posologie , Membrane synoviale/immunologie , Animaux , Méthotrexate/administration et posologie , Méthotrexate/pharmacologie , Petit ARN interférent/administration et posologie , Photothérapie dynamique/méthodes , Souris , Humains , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Cellules RAW 264.7 , Cytokines/métabolisme , Antirhumatismaux/administration et posologie , Microenvironnement cellulaire/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Photothérapie/méthodes , Peptides/administration et posologie
12.
Biomacromolecules ; 25(6): 3360-3372, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38771665

RÉSUMÉ

The simultaneous delivery of CpG oligonucleotide along with short interfering RNA (siRNA) has the potential to significantly boost the anticancer impact of siRNA medications. Our previous research demonstrated that Curdlan nanoparticles functionalized with adenosine are capable of selectively delivering therapeutic siRNA to cancerous cells through endocytosis mediated by adenosine receptors. Herein, we synthesized a dual-ligand-functionalized Curdlan polymer (denoted by CuMAN) to simultaneously target tumor cells and tumor-associated macrophages (TAMs). CuMAN nanoparticles containing CpG and siRNA demonstrated enhanced uptake by B16F10 tumor cells and bone marrow-derived macrophages, which are facilitated by AR on tumor cells and mannose receptor on macrophages. This led to increased release of pro-inflammatory cytokines in both in vitro and in vivo settings. The synergistic effect of CpG on TAMs and RNAi on tumor cells mediated by the CuMAN nanoparticle not only suppressed the tumor growth but also strongly inhibited the lung metastasis. Our findings indicate that the CuMAN nanoparticle has potential as an effective dual-targeting delivery system for nucleic acid therapeutics.


Sujet(s)
Nanoparticules , Petit ARN interférent , bêta-Glucanes , Animaux , bêta-Glucanes/composition chimique , bêta-Glucanes/pharmacologie , Petit ARN interférent/administration et posologie , Petit ARN interférent/pharmacologie , Petit ARN interférent/composition chimique , Nanoparticules/composition chimique , Souris , Souris de lignée C57BL , Oligodésoxyribonucléotides/composition chimique , Oligodésoxyribonucléotides/administration et posologie , Oligodésoxyribonucléotides/pharmacologie , Mélanome expérimental/anatomopathologie , Mélanome expérimental/traitement médicamenteux , Lignée cellulaire tumorale , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Ligands , Systèmes de délivrance de médicaments/méthodes , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques
13.
Hypertension ; 81(7): 1491-1499, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38690653

RÉSUMÉ

BACKGROUND: Small-interfering RNA (siRNA) targeting hepatic AGT (angiotensinogen) mRNA depletes AGT, lowering blood pressure for up to 6 months. However, certain situations may require a rapid angiotensin increase. The REVERSIR (RVR) - reverse siRNA silencing technology a potential approach to counteract siRNA effects. METHODS: Spontaneously hypertensive rats received 10 mg/kg AGT siRNA, and 3 weeks later were given AGT-RVR (1, 10, or 20 mg/kg). One week after AGT-RVR dosing, a redose of AGT siRNA assessed its post-AGT-RVR effectiveness for 2 weeks. Additionally, the impact of AGT-RVR after an equihypotensive dose of valsartan (4 mg/kg per day) was examined. RESULTS: Baseline mean arterial pressure (MAP) was 144±1 mm Hg. AGT siRNA reduced MAP by ≈16 mm Hg and AGT by >95%, while renin increased 25-fold. All AGT-RVR doses restored MAP to baseline within 4 to 7 days. Notably, 10 and 20 mg/kg restored AGT and renin to baseline, while 1 mg/kg allowed ≈50% AGT restoration, with renin remaining above baseline. A second AGT siRNA treatment, following 1 mg/kg AGT-RVR, reduced MAP to the same degree as the initial dose, while following 10 mg/kg AGT-RVR, it resulted in ≈50% of the first dose's MAP effect at 2 weeks. The valsartan-induced MAP reduction was unaffected by AGT-RVR. CONCLUSIONS: In spontaneously hypertensive rats, angiotensinogen-RVR dose-dependently reversed AGT siRNA-induced AGT reduction, normalizing MAP. MAP normalization persisted even with 50% recovered AGT levels, likely due to upregulated renin maintaining adequate angiotensin generation. Post-AGT-RVR dosing, a second AGT siRNA dose lowered MAP again.


Sujet(s)
Angiotensinogène , Antihypertenseurs , Hypertension artérielle , Petit ARN interférent , Rats de lignée SHR , Animaux , Angiotensinogène/génétique , Angiotensinogène/métabolisme , Petit ARN interférent/administration et posologie , Petit ARN interférent/pharmacologie , Petit ARN interférent/génétique , Rats , Hypertension artérielle/traitement médicamenteux , Hypertension artérielle/génétique , Hypertension artérielle/métabolisme , Antihypertenseurs/pharmacologie , Mâle , Pression sanguine/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Valsartan/pharmacologie , Système rénine-angiotensine/effets des médicaments et des substances chimiques
14.
Med ; 5(5): 383-385, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38733971

RÉSUMÉ

Hypertension is a modifiable risk factor for cardiovascular disease, the leading cause of death worldwide, yet most US adults with hypertension do not meet goal blood pressure. KARDIA-1 demonstrates the efficacy of zilebesiran, a subcutaneously administered small interfering RNA, for lowering blood pressure, presenting a novel treatment option for this deadly disease.1.


Sujet(s)
Hypertension artérielle , Petit ARN interférent , Hypertension artérielle/génétique , Humains , Petit ARN interférent/administration et posologie , Petit ARN interférent/usage thérapeutique , Pression sanguine/effets des médicaments et des substances chimiques
15.
Med Oncol ; 41(6): 149, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38739199

RÉSUMÉ

Because of the high biocompatibility, self-assembly capability, and CD71-mediated endocytosis, using human heavy chain ferritin (HFn) as a nanocarrier would greatly increase therapeutic effectiveness and reduce possible adverse events. Anti-PD-L1 siRNA can downregulate the level of PD-L1 on tumor cells, resulting in the activation of effector T cells against leukemia. Therefore, this study aimed to produce the tumor-targeting siPD-L1/HFn nanocarrier. Briefly, the HFn coding sequence was cloned into a pET-28a, and the constructed expression plasmid was subsequently transformed into E. coli BL21. After induction of Isopropyl ß-D-1-thiogalactopyranoside (IPTG), HFn was purified with Ni-affinity chromatography and dialyzed against PBS. The protein characteristics were analyzed using SDS-PAGE, Western Blot, and Dynamic light scattering (DLS). The final concentration was assessed using the Bicinchoninic acid (BCA) assay. The encapsulation was performed using the standard pH system. The treatment effects of siPD-L1/HFn were carried out on HL-60 and K-562 cancer cell lines. The RT-PCR was used to determine the mRNA expression of PD-L1. The biocompatibility and excretion of siPD-L1/HFn have also been evaluated. The expression and purity of HFn were well verified through SDS-PAGE, WB, and DLS. RT-PCR analyses also showed significant siRNA-mediated PD-L1 silencing in both HL-60 and K-562 cells. Our study suggested a promising approach for siRNA delivery. This efficient delivery system can pave the way for the co-delivery of siRNAs and multiple chemotherapies to address the emerging needs of cancer combination therapy.


Sujet(s)
Apoferritines , Antigène CD274 , Leucémie aigüe myéloïde , Petit ARN interférent , Humains , Petit ARN interférent/génétique , Petit ARN interférent/administration et posologie , Antigène CD274/métabolisme , Antigène CD274/génétique , Antigène CD274/antagonistes et inhibiteurs , Apoferritines/génétique , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/thérapie , Cellules HL-60 , Cellules K562 , Lignée cellulaire tumorale , Nanoparticules/composition chimique
16.
Nat Rev Drug Discov ; 23(6): 421-444, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38740953

RÉSUMÉ

RNA has sparked a revolution in modern medicine, with the potential to transform the way we treat diseases. Recent regulatory approvals, hundreds of new clinical trials, the emergence of CRISPR gene editing, and the effectiveness of mRNA vaccines in dramatic response to the COVID-19 pandemic have converged to create tremendous momentum and expectation. However, challenges with this relatively new class of drugs persist and require specialized knowledge and expertise to overcome. This Review explores shared strategies for developing RNA drug platforms, including layering technologies, addressing common biases and identifying gaps in understanding. It discusses the potential of RNA-based therapeutics to transform medicine, as well as the challenges associated with improving applicability, efficacy and safety profiles. Insights gained from RNA modalities such as antisense oligonucleotides (ASOs) and small interfering RNAs are used to identify important next steps for mRNA and gene editing technologies.


Sujet(s)
Oligonucléotides antisens , Humains , Oligonucléotides antisens/usage thérapeutique , Édition de gène/méthodes , COVID-19 , Traitements médicamenteux de la COVID-19 , Petit ARN interférent/usage thérapeutique , Petit ARN interférent/administration et posologie , Petit ARN interférent/génétique , SARS-CoV-2/génétique , SARS-CoV-2/effets des médicaments et des substances chimiques , ARN messager/génétique , Animaux , ARN/génétique
17.
Calcif Tissue Int ; 115(1): 85-96, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38733412

RÉSUMÉ

Autosomal dominant osteopetrosis type 2 (ADO2) is a rare inherited bone disorder characterised by dense but brittle bones. It displays striking phenotypic variability, with the most severe symptoms, including blindness and bone marrow failure. Disease management largely relies on symptomatic treatment since there is no safe and effective treatment. Most ADO2 cases are caused by heterozygous loss-of-function mutations in the CLCN7 gene, which encodes an essential Cl-/H+ antiporter for proper bone resorption by osteoclasts. Thus, siRNA-mediated silencing of the mutant allele is a promising therapeutic approach, but targeting bone for first-in-human translation remains challenging. Here, we demonstrate the utility of silicon-stabilised hybrid lipid nanoparticles (sshLNPs) as a next-generation nucleic acid nanocarrier capable of delivering allele-specific siRNA to bone. Using a Clcn7G213R knock-in mouse model recapitulating one of the most common human ADO2 mutations and based on the 129S genetic background (which produces the most severe disease phenotype amongst current models), we show substantial knockdown of the mutant allele in femur when siRNA targeting the pathogenic variant is delivered by sshLNPs. We observed lower areal bone mineral density in femur and reduced trabecular thickness in femur and tibia, when siRNA-loaded sshLNPs were administered subcutaneously (representing the most relevant administration route for clinical adoption and patient adherence). Importantly, sshLNPs have improved stability over conventional LNPs and enable 'post hoc loading' for point-of-care formulation. The treatment was well tolerated, suggesting that sshLNP-enabled gene therapy might allow successful clinical translation of essential new treatments for ADO2 and potentially other rare genetic bone diseases.


Sujet(s)
Allèles , Canaux chlorure , Nanoparticules , Ostéopétrose , Phénotype , Petit ARN interférent , Animaux , Canaux chlorure/génétique , Ostéopétrose/génétique , Ostéopétrose/thérapie , Souris , Petit ARN interférent/administration et posologie , Petit ARN interférent/génétique , Os et tissu osseux/métabolisme , Os et tissu osseux/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
18.
Expert Opin Drug Metab Toxicol ; 20(5): 399-406, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38706380

RÉSUMÉ

BACKGROUND: Methotrexate (MTX) is partially metabolized by aldehyde oxidase (AOX) in the liver and its clinical impact remains unclear. In this study, we aimed to demonstrate how AOX contributes to MTX-induced hepatotoxicity in vitro and clarify the relationship between concomitant AOX inhibitor use and MTX-associated liver injury development using the U.S. Food and Drug Administration Adverse Event Reporting System (FAERS). METHODS: We assessed intracellular MTX accumulation and cytotoxicity using HepG2 cells. We used the FAERS database to detect reporting odds ratio (ROR)-based MTX-related hepatotoxicity event signals. RESULTS: AOX inhibition by AOX inhibitor raloxifene and siRNA increased the MTX accumulation in HepG2 cells and enhanced the MTX-induced cell viability reduction. In the FAERS analysis, the ROR for MTX-related hepatotoxicity increased with non-overlap of 95% confidence interval when co-administered with drugs with higher Imax, u (maximum unbound plasma concentration)/IC50 (half-maximal inhibitory concentration for inhibition of AOX) calculated based on reported pharmacokinetic data. CONCLUSION: AOX inhibition contributed to MTX accumulation in the liver, resulting in increased hepatotoxicity. Our study raises concerns regarding MTX-related hepatotoxicity when co-administered with drugs that possibly inhibit AOX activity at clinical concentrations.


Sujet(s)
Systèmes de signalement des effets indésirables des médicaments , Aldehyde oxidase , Lésions hépatiques dues aux substances , Méthotrexate , Méthotrexate/effets indésirables , Méthotrexate/administration et posologie , Humains , Aldehyde oxidase/métabolisme , Lésions hépatiques dues aux substances/étiologie , Cellules HepG2 , Survie cellulaire/effets des médicaments et des substances chimiques , Antimétabolites antinéoplasiques/effets indésirables , Antimétabolites antinéoplasiques/administration et posologie , États-Unis , Food and Drug Administration (USA) , Petit ARN interférent/administration et posologie , Petit ARN interférent/pharmacologie , Concentration inhibitrice 50
19.
Chem Pharm Bull (Tokyo) ; 72(5): 512-517, 2024.
Article de Anglais | MEDLINE | ID: mdl-38811213

RÉSUMÉ

Cell-penetrating peptides (CPPs) serve as potent vehicles for delivering membrane-impermeable compounds, including nucleic acids, into cells. In a previous study, we reported the successful intracellular delivery of small interfering RNAs (siRNAs) with negligible cytotoxicity using a peptide containing an unnatural amino acid (dipropylglycine). In the present study, we employed the same seven peptides as the previous study to evaluate their efficacy in delivering plasmid DNA (pDNA) intracellularly. Although pDNA and siRNA are nucleic acids, they differ in size and biological function, which may influence the optimal peptide sequences for their delivery. Herein, three peptides demonstrated effective pDNA transfection abilities. Notably, only one of the three peptides previously exhibited efficient gene-silencing effect with siRNA. These findings validate our hypothesis and offer insights for the personalized design of CPPs for the delivery of pDNA and siRNA.


Sujet(s)
Peptides de pénétration cellulaire , ADN , Plasmides , Petit ARN interférent , Peptides de pénétration cellulaire/composition chimique , Humains , ADN/composition chimique , Petit ARN interférent/composition chimique , Petit ARN interférent/administration et posologie , Glycine/composition chimique , Transfection , Cellules HeLa , Survie cellulaire/effets des médicaments et des substances chimiques
20.
Int J Pharm ; 658: 124199, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38703928

RÉSUMÉ

Dendrimers have emerged as an important group of nanoparticles to transport drugs, DNA, or RNA into target cells in cancer and other diseases. Various functional modifications can be imposed on dendrimers to increase the efficacy and specificity in delivering their cargo to the target cells and decrease their toxicity. In the present work, we evaluated the potential of carbosilane polyphenolic dendrimers modified with caffeic acid (CA) and polyethylene glycol (PEG) to deliver proapoptotic Mcl-1 and Bcl-2 siRNAs to A549 cancer cells. Dendrimers formed stable complexes with siRNAs as assessed by transmission electron microscopy and gel electrophoresis. Modification of dendrimers with PEG reduced the size and the zeta potential of dendrimer/siRNA complexes. The presence of PEG caused a red shift of the CD spectrum, and this effect was the more pronounced, the higher the dendrimer/siRNA ratio was. The nanocomplexes were internalized by A549. All studied dendrimer/siRNA formulations inhibited tumor cell migration and adhesion and caused an increase in the population of early apoptotic cells. Among four tested dendrimers, the polyphenolic compound containing two caffeic acid moieties complexed with siRNA demonstrated the lowest polydispersity index and showed an excellent transfection profile. In conclusion, this dendrimer are a promising candidate for the delivery of siRNA into cancer cells in further in vivo studies.


Sujet(s)
Apoptose , Dendrimères , Polyéthylène glycols , Polyphénols , Petit ARN interférent , Humains , Dendrimères/composition chimique , Dendrimères/administration et posologie , Petit ARN interférent/administration et posologie , Petit ARN interférent/composition chimique , Cellules A549 , Apoptose/effets des médicaments et des substances chimiques , Polyphénols/composition chimique , Polyphénols/pharmacologie , Polyphénols/administration et posologie , Polyéthylène glycols/composition chimique , Protéines proto-oncogènes c-bcl-2/génétique , Acides caféiques/composition chimique , Acides caféiques/pharmacologie , Acides caféiques/administration et posologie , Protéine Mcl-1/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Vecteurs de médicaments/composition chimique , Silanes/composition chimique , Transfection/méthodes , Lignée cellulaire tumorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...