Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.073
Filter
1.
Pharm Pat Anal ; 13(1-3): 31-43, 2024.
Article in English | MEDLINE | ID: mdl-39324857

ABSTRACT

The release of active agents in tumors rather than normal tissues, limits systemic exposure and toxicities. Targeting over-expressed esterase enzyme in the tumor microenvironment can selectively release immune-active agents like Programmed Death-1 (PD-1) and PD-1 ligand inhibitors from ester-sensitive lipid nanocarriers, offering a novel approach compared with conventional therapies. PD-1 and PD-L1 association cause T-cell inactivation, whereas blocking their association improves their cytotoxic mechanism. The patent application US2022/0080051-A1 discloses a novel immune-active agent conjugated with lipid to form a nanocarrier for esterase-sensitive release. These nanocarriers selectively enter leaky vasculature of tumors through enhanced permeability and retention effect, undergo ester cleavage to release agents, and are reported to increase bioavailability by 24 times. Further, with other agents or alone it achieves targeted synergistic cancer therapy. Also, the current patent spotlight delves into the crucial formulation considerations necessary for obtaining successful approval of lipidic nano products from relevant regulatory authorities.


[Box: see text].


Subject(s)
Antineoplastic Agents , Drug Carriers , Esterases , Lipids , Nanoparticles , Humans , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Lipids/chemistry , Lipids/administration & dosage , Esterases/metabolism , Animals , Patents as Topic , Neoplasms/drug therapy , Drug Liberation
2.
Otol Neurotol ; 45(9): 1068-1077, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39165134

ABSTRACT

HYPOTHESIS: Microneedle-mediated intracochlear injection of siRNA-Lipofectamine through the round window membrane (RWM) can be used to transfect cells within the cochlea. BACKGROUND: Our laboratory has developed 100-µm diameter hollow microneedles for intracochlear injection through the guinea pig RWM. In this study, we test the feasibility of microneedle-mediated injection of siRNA and Lipofectamine, a commonly used reagent with known cellular toxicity, through the RWM for cochlear transfection. METHODS: Fluorescently labeled scramble siRNA was diluted into Lipofectamine RNAiMax and OptiMEM. One microliter of 5 µM siRNA was injected through the RWM of Hartley guinea pigs at a rate of 1 µl/min (n = 22). In a control group, 1.0 µl of Lipofectamine, with no siRNA, was diluted into OptiMEM and injected in a similar fashion (n = 5). Hearing tests were performed before and either at 24 hours, 48 hours, or 5 days after injection. Afterward, animals were euthanized, and cochleae were harvested for imaging. Control cochleae were processed in parallel to untreated guinea pigs. RESULTS: Fluorescence, indicating successful transfection, was observed within the basal and middle turns of the cochlea with limited distribution in the apex at 24 and 48 hours. Signal was most intense in the organ of Corti, spiral ligament, and spiral ganglion. Little to no fluorescence was observed at 5 days post-injection. No significant changes in auditory brainstem response (ABR) were noted post-perforation at 5 days, suggesting that siRNA-Lipofectamine at low doses does not cause cochlear toxicity. CONCLUSIONS: Small volumes of siRNA and Lipofectamine can be effectively delivered to cochlear structures using microneedles, paving the way for atraumatic cochlear gene therapy.


Subject(s)
Genetic Therapy , Liposomes , RNA, Small Interfering , Transfection , Animals , Guinea Pigs , RNA, Small Interfering/administration & dosage , Transfection/methods , Genetic Therapy/methods , Lipids/administration & dosage , Lipids/chemistry , Cochlea , Round Window, Ear , Needles , Evoked Potentials, Auditory, Brain Stem/drug effects , Ear, Inner , Microinjections/methods
3.
J Control Release ; 374: 550-562, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146981

ABSTRACT

mRNA incorporated in lipid nanoparticles (LNPs) became a new class of vaccine modality for induction of immunity against COVID-19 and ushered in a new era in vaccine development. Here, we report a novel, easy-to-execute, and cost effective engineered extracellular vesicles (EVs)-based combined mRNA and protein vaccine platform (EVX-M+P vaccine) and explore its utility in proof-of-concept immunity studies in the settings of cancer and infectious disease. As a first example, we engineered EVs, natural nanoparticle carriers shed by all cells, to contain ovalbumin mRNA and protein (EVOvaM+P vaccine) to serve as cancer vaccine against ovalbumin-expressing melanoma tumors. EVOvaM+P administration to mice with established melanoma tumors resulted in tumor regression associated with effective humoral and adaptive immune responses. As a second example, we generated engineered EVs that contain Spike (S) mRNA and protein to serve as a combined mRNA and protein vaccine (EVSpikeM+P vaccine) against SARS-CoV-2 infection. EVSpikeM+P vaccine administration in mice and baboons elicited robust production of neutralizing IgG antibodies against RBD (receptor binding domain) of S protein and S protein specific T cell responses. Our proof-of-concept study describes a new platform with an ability for rapid development of combination mRNA and protein vaccines employing EVs for deployment against cancer and other diseases.


Subject(s)
COVID-19 Vaccines , COVID-19 , Cancer Vaccines , Extracellular Vesicles , Mice, Inbred C57BL , Nanoparticles , Ovalbumin , RNA, Messenger , Animals , Extracellular Vesicles/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , RNA, Messenger/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , Ovalbumin/immunology , Ovalbumin/administration & dosage , Mice , Female , Nanoparticles/administration & dosage , Nanoparticles/chemistry , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Humans , Cell Line, Tumor , Melanoma/immunology , Melanoma/therapy , Lipids/chemistry , Lipids/administration & dosage , Liposomes
4.
Expert Opin Drug Deliv ; 21(8): 1191-1211, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39172249

ABSTRACT

INTRODUCTION: Wounds, resulting from traumas, surgery, burns or diabetes, are important medical problems due to the complexity of wound healing process regarding healing times and healthcare costs. Nanosystems have emerged as promising candidates in this field thank to their properties and versatile applications in drugs delivery. AREAS COVERED: Lipid-based nanosystems (LBN) are described for wound treatment, highlighting their different behaviors when interacting with the cutaneous tissue. The role of nanosystems in delivering mostly natural compounds on skin as well as the technological and engineering strategies to increase their efficiency in wound healing effect are reviewed. Finally, in vitro, ex-vivo and in vivo studies are reported. EXPERT OPINION: LBN have shown promise in addressing the challenges of wound healing as they can improve the stability of drugs used in wound therapy, leading to higher efficacy and fewer adverse effects as compared to traditional formulations. LBNs being involved in the inflammatory and proliferation stages of the wound healing process, enable the modification of wound healing through multiple ways. In addition, the use of new technologies, including 3D bioprinting and photobiomodulation, may lead to potential breakthroughs in wound healing. This would provide clinicians with more potent forms of therapy for wound healing.


Subject(s)
Drug Delivery Systems , Lipids , Wound Healing , Wound Healing/drug effects , Humans , Animals , Lipids/chemistry , Lipids/administration & dosage , Skin/drug effects , Nanoparticles/chemistry , Wounds and Injuries/drug therapy , Wounds and Injuries/therapy , Drug Stability
5.
J Control Release ; 374: 590-605, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39208936

ABSTRACT

Herein, we reported novel docetaxel-decorated solid lipid nanoparticle (DCT-SLN)-loaded dual thermoreversible system (DCT-DRTS) for intramuscular administration with reduced burst effect, sustained release and improved antitumor efficacy. The optimized DCT-DRTs was subjected to in-vitro and in-vivo analyses. Antitumor evaluation of the DCT-DRTS was executed and compared with DCT-hydrogel, and DCT-suspension trailed by the histopathological and immune-histochemical analyses. The DCT-SLN gave a mean particle size of 157 nm and entrapment efficiency of 93 %. It was a solid at room temperature, and changed to liquid at physiological temperature due to its melting point of about 32 °C. Unlikely, poloxamer mixture remained liquefied at 25-27 °C, however converted to gel at physiological temperature. This behavior demonstrated opposed reversible property of the DCT-SLN and poloxamer hydrogel in DCT-DRTS system, making it ideal for intramuscular administration and quick gelation inside the body. The DCT-DRTS sustained the drugs release and unlike DCT-hydrogel, the preliminary plasma concentration of DCT-DRTS was significantly reduced, overcoming the burst release. A meaningfully enhanced antitumor efficacy and improved survival rate was observed from DCT-DRTS in tumor cell xenograft athymic nude mice. Additionally, increased apoptotic and reduced proliferation markers were observed in DCT-DRTS treated tumor masses. It was concluded that DCT-DRTS may be a suitable choice for intramuscular administration of DCT with sustained release, improved bioavailability, reduced toxicity and enhanced antitumor effects.


Subject(s)
Antineoplastic Agents , Delayed-Action Preparations , Docetaxel , Hydrogels , Nanoparticles , Animals , Hydrogels/chemistry , Hydrogels/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Humans , Injections, Intramuscular , Docetaxel/administration & dosage , Docetaxel/pharmacokinetics , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Delayed-Action Preparations/chemistry , Mice, Inbred BALB C , Cell Line, Tumor , Drug Liberation , Temperature , Mice, Nude , Poloxamer/chemistry , Mice , Drug Delivery Systems , Female , Lipids/chemistry , Lipids/administration & dosage , Male , Drug Carriers/chemistry , Neoplasms/drug therapy , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Taxoids/chemistry , Liposomes
6.
Int J Pharm ; 662: 124464, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39033939

ABSTRACT

Leishmaniases, a group of neglected tropical diseases caused by an intracellular parasite of the genus Leishmania, have significant impacts on global health. Current treatment options are limited due to drug resistance, toxicity, and high cost. This study aimed to develop nanostructured lipid carriers (NLCs) for delivering Citrus sinensis essential oil (CSEO) and its main constituent, R-limonene, against leishmaniasis. The influence of surface-modified NLCs using chitosan was also examined. The NLCs were prepared using a warm microemulsion method, and surface modification with chitosan was achieved through electrostatic interaction. These nanocarriers were characterized by differential scanning calorimetry (DSC), X-ray diffraction (XRD), transmission electron microscopy, and dynamic light scattering (DLS). In vitro cytotoxicity was assessed in L929 and RAW 264.7 cells, and leishmanicidal activity was evaluated against promastigote and amastigote forms. The NLCs were spherical, with particle sizes ranging from 97.9 nm to 111.3 nm. Chitosan-coated NLCs had a positive surface charge, with zeta potential values ranging from 45.8 mV to 59.0 mV. Exposure of L929 cells to NLCs resulted in over 70 % cell viability. Conversely, surface modification significantly reduced the viability of promastigotes (93 %) compared to free compounds. Moreover, chitosan-coated NLCs presented a better IC50 against the amastigote forms than uncoated NLCs. Taken together, these findings demonstrate the feasibility of using NLCs to overcome the limitations of current leishmaniasis treatments, warranting further research.


Subject(s)
Cell Survival , Chitosan , Citrus sinensis , Drug Carriers , Limonene , Lipids , Nanoparticles , Oils, Volatile , Oils, Volatile/administration & dosage , Oils, Volatile/chemistry , Oils, Volatile/pharmacology , Animals , Mice , Limonene/chemistry , Limonene/administration & dosage , Limonene/pharmacology , Drug Carriers/chemistry , RAW 264.7 Cells , Cell Survival/drug effects , Chitosan/chemistry , Chitosan/administration & dosage , Lipids/chemistry , Lipids/administration & dosage , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Citrus sinensis/chemistry , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/chemistry , Leishmaniasis/drug therapy , Particle Size , Cell Line , Leishmania/drug effects , Terpenes/chemistry , Terpenes/pharmacology , Terpenes/administration & dosage , Nanostructures/chemistry , Nanostructures/administration & dosage
7.
Int J Pharm ; 660: 124300, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38851409

ABSTRACT

Uveal melanoma is one of the most common and aggressive intraocular malignancies, and, due to its great capability of metastasize, it constitutes the most incident intraocular tumor in adults. However, to date there is no effective treatment since achieving the inner ocular tissues still constitutes one of the greatest challenges in actual medicine, because of the complex structure and barriers. Uncoated and PEGylated nanostructured lipid carriers were developed to achieve physico-chemical properties (mean particle size, homogeneity, zeta potential, pH and osmolality) compatible for the ophthalmic administration of (S)-(-)-MRJF22, a new custom-synthetized prodrug for the potential treatment of uveal melanoma. The colloidal physical stability was investigated at different temperatures by Turbiscan® Ageing Station. Morphology analysis and mucoadhesive studies highlighted the presence of small particles suitable to be topically administered on the ocular surface. In vitro release studies performed using Franz diffusion cells demonstrated that the systems were able to provide a slow and prolonged prodrug release. In vitro cytotoxicity test on Human Corneal Epithelium and Human Uveal Melanoma cell lines and Hen's egg-chorioallantoic membrane test showed a dose-dependent cytotoxic effect of the free prodrug on corneal cells, whose cytocompatibility improved when encapsulated into nanoparticles, as also confirmed by in vivo studies on New Zealand albino rabbits. Antiangiogenic capability and preventive anti-inflammatory properties were also investigated on embryonated eggs and rabbits, respectively. Furthermore, preliminary in vivo biodistribution images of fluorescent nanoparticles after topical instillation in rabbits' eyes, suggested their ability to reach the posterior segment of the eye, as a promising strategy for the treatment of choroidal uveal melanoma.


Subject(s)
Administration, Ophthalmic , Chorioallantoic Membrane , Drug Carriers , Melanoma , Nanoparticles , Prodrugs , Uveal Neoplasms , Uveal Neoplasms/drug therapy , Uveal Neoplasms/pathology , Melanoma/drug therapy , Melanoma/pathology , Animals , Humans , Rabbits , Cell Line, Tumor , Chorioallantoic Membrane/drug effects , Drug Carriers/chemistry , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Prodrugs/administration & dosage , Prodrugs/chemistry , Lipids/chemistry , Lipids/administration & dosage , Drug Liberation , Cell Survival/drug effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Chick Embryo , Epithelium, Corneal/drug effects , Particle Size
8.
J Control Release ; 372: 168-175, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38844178

ABSTRACT

Influenza outbreaks are a major burden worldwide annually. While seasonal vaccines do provide protection against infection, they are limited in that they need to be updated every year to account for the constantly mutating virus. Recently, lipid nanoparticles (LNPs) encapsulating mRNA have seen major success as a vaccine platform for SARS-CoV-2. Herein, we applied LNPs to deliver an mRNA encoding a computationally optimized broadly active (COBRA) influenza immunogen. These COBRA mRNA LNPs induced a broadly active neutralizing antibody response and protection after lethal influenza challenge. To further increase the immunogenicity of the COBRA mRNA LNPs, we combined them with acetalated dextran microparticles encapsulating a STING agonist. Contrary to recent findings, the STING agonist decreased the immunogenicity of the COBRA mRNA LNPs which was likely due to a decrease in mRNA translation as shown in vitro. Overall, this work aids in future selection of adjuvants to use with mRNA LNP vaccines.


Subject(s)
Influenza Vaccines , Nanovaccines , Nucleotides, Cyclic , Animals , Female , Mice , Adjuvants, Immunologic/administration & dosage , Antibodies, Neutralizing/immunology , Dextrans/chemistry , Dextrans/administration & dosage , Immunogenicity, Vaccine , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Lipids/chemistry , Lipids/administration & dosage , Liposomes , Mice, Inbred BALB C , mRNA Vaccines , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanovaccines/administration & dosage , Nanovaccines/chemistry , Nucleotides, Cyclic/administration & dosage , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Polymers/chemistry , Polymers/administration & dosage , RNA, Messenger/administration & dosage , RNA, Messenger/immunology
9.
J Control Release ; 372: 417-432, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908758

ABSTRACT

This study introduces the nanobromhexine lipid particle (NBL) platform designed for effective pulmonary drug delivery. Inspired by respiratory virus transport mechanisms, NBL address challenges associated with mucus permeation and inflammation in pulmonary diseases. Composed of low molecular weight polyethylene glycol-coated lipid nanoparticles with bromhexine hydrochloride, NBL exhibit a size of 118 ± 24 nm, a neutral zeta potential, osmolarity of 358 ± 28 mOsmol/kg, and a pH of 6.5. Nebulizing without leakage and showing no toxicity to epithelial cells, NBL display mucoadhesive properties with a 60% mucin-binding efficiency. They effectively traverse the dense mucus layer of Calu-3 cultures in an air-liquid interface, as supported by a 55% decrease in MUC5AC density and a 29% increase in nanoparticles internalization compared to non-exposed cells. In assessing immunomodulatory effects, NBL treatment in SARS-CoV-2-infected lung cells leads to a 40-fold increase in anti-inflammatory MUC1 gene expression, a proportional reduction in pro-inflammatory IL-6 expression, and elevated anti-inflammatory IL-10 expression. These findings suggest a potential mechanism to regulate the excessive IL-6 expression triggered by virus infection. Therefore, the NBL platform demonstrates promising potential for efficient pulmonary drug delivery and immunomodulation, offering a novel approach to addressing mucus permeation and inflammation in pulmonary diseases.


Subject(s)
Lung , Nanoparticles , SARS-CoV-2 , Nanoparticles/administration & dosage , Humans , Lung/metabolism , SARS-CoV-2/drug effects , Drug Delivery Systems , Immunomodulation , Cell Line , Mucin-1/metabolism , COVID-19 , Lipids/chemistry , Lipids/administration & dosage , Mucus/metabolism , Polyethylene Glycols/chemistry , Epithelial Cells/metabolism , Epithelial Cells/drug effects , COVID-19 Drug Treatment , Mucin 5AC/metabolism , Liposomes
10.
J Control Release ; 372: 778-794, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936744

ABSTRACT

Alopecia areata affects over 140 million people worldwide and causes severe psychological distress. The Janus kinase (JAK) inhibitor, tofacitinib, shows significant potential in therapeutic applications for treating alopecia areata; however, the systemic adverse effects of oral administration and low absorption rate at the target site limit its application. Hence, to address this issue, we designed topical formulations of tofacitinib-loaded cationic lipid nanoparticles (TFB-cNLPs) with particle sizes of approximately 200 nm. TFB-cNLPs promoted percutaneous absorption and hair follicle targeting in an ex vivo pig ear model. TFB-cNLP decreased IFN-γ-induced alopecia areata symptoms in an in vitro follicle model by blocking the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. It also reduced the number of CD8+NKG2D+T cells in a C3H mouse model of alopecia areata in vivo, thereby inhibiting the progression of alopecia areata and reversing hair loss. These findings suggest that TFB-cNLP enhanced hair follicle targeting and has the potential for topical treatment or prevention of alopecia areata.


Subject(s)
Alopecia Areata , Drug Carriers , Hair Follicle , Lipids , Piperidines , Pyrimidines , Skin Absorption , Animals , Alopecia Areata/drug therapy , Hair Follicle/metabolism , Hair Follicle/drug effects , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Piperidines/pharmacology , Piperidines/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Swine , Lipids/chemistry , Lipids/administration & dosage , Drug Carriers/chemistry , Mice, Inbred C3H , Nanoparticles/administration & dosage , Mice , Nanostructures/administration & dosage , Female , Liposomes
11.
J Control Release ; 372: 609-618, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942082

ABSTRACT

Therapeutically manipulating the stimulator of interferon genes (STING) pathway has promising potential for enhancing antitumor immunity. Agonists of this pathway (STING agonists) are being evaluated in clinical trials. Loading the STING agonists into lipid nanoparticles (LNPs) increases their safety and efficacy. We previously developed STING agonists loaded LNPs consisting of the ionizable lipid YSK12-C4 (YSK12-LNPs), which showed significant antitumor effects. However, it is largely unclear how the in vivo fate of STING agonists loaded LNPs affects the antitumor immune responses. In this study, we compared the YSK12-LNPs with LNPs composed of DLin-MC3-DMA (MC3-LNPs) showing different in vivo fates. Biodistribution and flow cytometry analyses of mouse tissues revealed that the MC3-LNPs delivered higher amounts of STING agonists to the liver than the YSK12-LNPs. Additionally, significantly more liver leukocytes internalized the MC3-LNPs than the YSK12-LNPs. In contrast, the YSK12-LNPs delivered higher amounts of STING agonists to the liver leukocytes than the MC3-LNPs, leading to the effective induction of innate immunity and inflammation in the tumors. However, the antitumor effects in the B16-F10 lung metastasis and CT26 tumor models were comparable. Interestingly, flow cytometry analyses suggested that the YSK12-LNPs were more likely to activate natural killer cells and M1 macrophages, while the MC3-LNPs were more likely to activate CD8+ T cells. Our data suggest that different antitumor immune response mechanisms may operate depending on the characteristics and distribution of the LNPs.


Subject(s)
Lipids , Membrane Proteins , Mice, Inbred C57BL , Nanoparticles , Animals , Nanoparticles/administration & dosage , Lipids/chemistry , Lipids/administration & dosage , Female , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Tissue Distribution , Immunity, Innate/drug effects , Mice , Liver/metabolism , Liver/drug effects , Neoplasms/drug therapy , Neoplasms/immunology , Liposomes
12.
Biomol Biomed ; 24(5): 1361-1369, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-38696705

ABSTRACT

Bile leakage is a common complication following liver surgeries, particularly in the cases of liver hydatid cyst operations. Currently, there is no adequate method which could be utilized to prevent this complication effectively. Our study aimed to assess the efficacy of the biliary lipid test (BLT) in reducing biliary complications after hydatid cyst surgery. We retrospectively included patients who underwent open liver hydatid cyst surgery between January 2011 and January 2024. The study encompassed 122 patients, with 41 males and 81 females, ranging in age from 18 to 79 years. In the BLT group, a lipid solution was injected transcystically after cholecystectomy. The BLT was performed on 65 patients, while 57 patients did not undergo the test. Following the transcystic injection of the lipid solution, orifices at the site of lipid droplets that became visible were ligated with 5.0 prolene sutures. A total of 200 leak sites were sutured. Remarkably, none of the patients in the BLT group experienced postoperative bile leakage lasting longer than one week. Consequently, a shorter hospital stay was observed in this group. Transcystic injection of the lipid solution with distal clamping effectively demonstrated leak sites, and suturing these sites prevented postoperative bile leakage. Our study demonstrates the effectiveness of the LIpid test in LIver Hydatid Cyst Surgery (Lili-Hics) in reducing biliary complications following hydatid cyst surgery.


Subject(s)
Echinococcosis, Hepatic , Lipids , Humans , Male , Female , Middle Aged , Echinococcosis, Hepatic/surgery , Adult , Aged , Adolescent , Lipids/administration & dosage , Retrospective Studies , Young Adult , Postoperative Complications/prevention & control , Bile/metabolism
13.
Int J Pharm ; 658: 124222, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38735632

ABSTRACT

Dry eye disease (DED) is a chronic multifactorial disorder of the ocular surface caused by tear film dysfunction and constitutes one of the most common ocular conditions worldwide. However, its treatment remains unsatisfactory. While artificial tears are commonly used to moisturize the ocular surface, they do not address the underlying causes of DED. Apigenin (APG) is a natural product with anti-inflammatory properties, but its low solubility and bioavailability limit its efficacy. Therefore, a novel formulation of APG loaded into biodegradable and biocompatible nanoparticles (APG-NLC) was developed to overcome the restricted APG stability, improve its therapeutic efficacy, and prolong its retention time on the ocular surface by extending its release. APG-NLC optimization, characterization, biopharmaceutical properties and therapeutic efficacy were evaluated. The optimized APG-NLC exhibited an average particle size below 200 nm, a positive surface charge, and an encapsulation efficiency over 99 %. APG-NLC exhibited sustained release of APG, and stability studies demonstrated that the formulation retained its integrity for over 25 months. In vitro and in vivo ocular tolerance studies indicated that APG-NLC did not cause any irritation, rendering them suitable for ocular topical administration. Furthermore, APG-NLC showed non-toxicity in an epithelial corneal cell line and exhibited fast cell internalization. Therapeutic benefits were demonstrated using an in vivo model of DED, where APG-NLC effectively reversed DED by reducing ocular surface cellular damage and increasing tear volume. Anti-inflammatory assays in vivo also showcased its potential to treat and prevent ocular inflammation, particularly relevant in DED patients. Hence, APG-NLC represent a promising system for the treatment and prevention of DED and its associated inflammation.


Subject(s)
Apigenin , Drug Carriers , Dry Eye Syndromes , Lipids , Nanoparticles , Animals , Apigenin/administration & dosage , Apigenin/chemistry , Apigenin/pharmacology , Apigenin/pharmacokinetics , Drug Carriers/chemistry , Dry Eye Syndromes/drug therapy , Humans , Rabbits , Lipids/chemistry , Lipids/administration & dosage , Cell Line , Nanoparticles/chemistry , Administration, Ophthalmic , Drug Liberation , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/pharmacokinetics , Particle Size , Nanostructures/administration & dosage , Nanostructures/chemistry , Male
14.
Cancer Res ; 84(14): 2352-2363, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38718316

ABSTRACT

Pulmonary delivery of immunostimulatory agents such as poly(I:C) to activate double-stranded RNA sensors MDA5 and RIG-I within lung-resident antigen-presenting cells is a potential strategy to enhance antitumor immunity by promoting type I interferon secretion. Nevertheless, following pulmonary delivery, poly(I:C) suffers from rapid degradation and poor endosomal escape, thus limiting its potency. Inspired by the structure of a virus that utilizes internal viral proteins to tune the loading and cytosolic delivery of viral nucleic acids, we developed a liponanogel (LNG)-based platform to overcome the delivery challenges of poly(I:C). The LNG comprised an anionic polymer hyaluronic acid-based nanogel core coated by a lipid shell, which served as a protective layer to stabilize the nanogel core in the lungs. The nanogel core was protonated within acidic endosomes to enhance the endosomal membrane permeability and cytosolic delivery of poly(I:C). After pulmonary delivery, LNG-poly(I:C) induced 13.7-fold more IFNß than poly(I:C) alone and two-fold more than poly(I:C) loaded in the state-of-art lipid nanoparticles [LNP-poly(I:C)]. Additionally, LNG-poly(I:C) induced more potent CD8+ T-cell immunity and stronger therapeutic effects than LNP-poly(I:C). The combination of LNG-poly(I:C) and PD-L1 targeting led to regression of established lung metastases. Due to the ease of manufacturing and the high biocompatibility of LNG, pulmonary delivery of LNG may be broadly applicable to the treatment of different lung tumors and may spur the development of innovative strategies for cancer immunotherapy. Significance: Pulmonary delivery of poly(I:C) with a virus-inspired inhalable liponanogel strongly activates cytosolic MDA5 and RIG-I and stimulates antitumor immunity, representing a promising strategy for safe and effective treatment of metastatic lung tumors.


Subject(s)
Lung Neoplasms , Poly I-C , Lung Neoplasms/secondary , Lung Neoplasms/immunology , Lung Neoplasms/drug therapy , Animals , Mice , Poly I-C/administration & dosage , Humans , Mice, Inbred C57BL , Nanogels/chemistry , Cell Line, Tumor , Female , Administration, Inhalation , Lipids/chemistry , Lipids/administration & dosage
15.
J Control Release ; 371: 179-192, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795814

ABSTRACT

The delivery of vaccines plays a pivotal role in influencing the strength and longevity of the immune response and controlling reactogenicity. Mucosal immunization, as compared to parenteral vaccination, could offer greater protection against respiratory infections while being less invasive. While oral vaccination has been presumed less effective and believed to target mainly the gastrointestinal tract, trans-buccal delivery using mucoadhesive films (MAF) may allow targeted delivery to the mucosa. Here we present an effective strategy for mucosal delivery of several vaccine platforms incorporated in MAF, including DNA plasmids, viral vectors, and lipid nanoparticles incorporating mRNA (mRNA/LNP). The mRNA/LNP vaccine formulation targeting SARS-CoV-2 as a proof of concept remained stable within MAF consisting of slowly releasing water-soluble polymers and an impermeable backing layer, facilitating enhanced penetration into the oral mucosa. This formulation elicited antibody and cellular responses comparable to the intramuscular injection, but also induced the production of mucosal IgAs, highlighting its efficacy, particularly for use as a booster vaccine and the potential advantage for protection against respiratory infections. The MAF vaccine preparation demonstrates significant advantages, such as efficient delivery, stability, and simple noninvasive administration with the potential to alleviate vaccine hesitancy.


Subject(s)
COVID-19 Vaccines , Nanoparticles , Animals , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Administration, Oral , Nanoparticles/administration & dosage , Mouth Mucosa/immunology , COVID-19/prevention & control , Female , Mice, Inbred BALB C , SARS-CoV-2/immunology , Mice , Drug Delivery Systems/methods , Humans , Lipids/chemistry , Lipids/administration & dosage , RNA, Messenger/administration & dosage , Liposomes
16.
Int J Pharm ; 660: 124172, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38679243

ABSTRACT

The goal of this study was to formulate tacrolimus nanogel based on nanostructured lipid carrier (NLC) in order to improve the efficacy, aesthetic, and patient compliance for the treatment of psoriasis. The microemulsion method was used to create phase diagrams and NLCs were prepared using points obtained from the microemulsion region and characterized. The gelling agent carbopol was used to develop an NLC-based nanogel. The pH, drug assay, viscosity, spreadability, and in vitro release of the nanogel, were evaluated. Ex vivo cytotoxicity of the formulation was assessed in murine fibroblast cells. Oxazolone and imiquimod models of psoriasis were used to assess the effectiveness of the nanogel. The NLCs exhibited a submicron particle size of 320 ± 10 nm, a low polydispersity index (<0.3), and a zeta potential of -19.4 mV. Morphological analysis revealed spherical nanoparticles with an encapsulation efficiency of 60 ± 3 %. The nanogel maintained a pH of 6.0 ± 0.5 and possessed a remarkable drug content of 99.73 ± 1.4 %. It exhibited pseudoplastic flow behaviour, ensuring easy spreadability, and demonstrated sustained drug release exceeding 90 % over a 24-hr period. Ex vivo cytotoxicity assessments revealed that the nanogel was safe because no cell death was induced. Nanogel resolved psoriatic blisters, was non-irritating and improved skin elasticity. The favorable properties, safety profile, and remarkable efficacy show the potential of the nanogel as a patient-friendly and effective therapeutic option for psoriasis treatment.


Subject(s)
Drug Carriers , Drug Liberation , Lipids , Nanogels , Psoriasis , Tacrolimus , Psoriasis/drug therapy , Animals , Drug Carriers/chemistry , Mice , Lipids/chemistry , Lipids/administration & dosage , Tacrolimus/administration & dosage , Tacrolimus/chemistry , Tacrolimus/pharmacokinetics , Nanogels/chemistry , Delayed-Action Preparations , Particle Size , Nanostructures/chemistry , Nanostructures/administration & dosage , Nanoparticles/chemistry , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/chemistry , Male , Imiquimod/administration & dosage , Fibroblasts/drug effects , Chemistry, Pharmaceutical/methods , Gels , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Polyethyleneimine
17.
Drug Deliv Transl Res ; 14(10): 2615-2628, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38587758

ABSTRACT

Fabry disease (FD) results from a lack of activity of the lysosomal enzyme α-Galactosidase A (α-Gal A), leading to the accumulation of glycosphingolipids in several different cell types. Protein supplementation by pDNA or mRNA delivery presents a promising strategy to tackle the underlying genetic defect in FD. Protein-coding nucleic acids in FD can be either delivered to the most affected sites by the disease, including heart, kidney and brain, or to specialized organs that can act as a production factory of the enzyme, such as the liver. Lipid-based systems are currently at the top of the ranking of non-viral nucleic acid delivery systems, and their versatility allows the linking to the surface of a wide range of molecules to control their biodistribution after intravenous administration. This systematic review follows the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) statement guidelines and provides an overview and discussion of the targeting ligands that have been employed so far to actively vectorize intravenously administered non-viral vectors based on lipid carriers to clinically relevant organs in the treatment of FD, for protein-coding nucleic acid (pDNA and mRNA) supplementation. Among the thirty-two studies included, the majority focus on targeting the liver and brain. The targeting of the heart has been reported to a lesser degree, whereas no articles addressing kidney-targeting have been recorded. Although a great effort has been made to develop organ-specific nucleic acid delivery systems, the design of active-targeted carriers with high quality, good clinical translation, and large-scale manufacturing capacity is still challenging.


Subject(s)
Fabry Disease , Lipids , Fabry Disease/therapy , Fabry Disease/drug therapy , Humans , Animals , Lipids/chemistry , Lipids/administration & dosage , alpha-Galactosidase/administration & dosage , RNA, Messenger/administration & dosage , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Drug Delivery Systems
18.
Drug Deliv Transl Res ; 14(10): 2917-2929, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38615156

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a highly prevalent chronic liver disease that can progress to end-stage conditions with life-threatening complications, but no pharmacologic therapy has been approved. Drug delivery systems such as lipid nanocapsules (LNC) are very versatile platforms that are easy to produce and can induce the secretion of the native glucagon-like peptide 1 (GLP-1) when orally administered. GLP-1 analogs are currently being studied in clinical trials in the context of MASLD. Our nanosystem provides with increased levels of the native GLP-1 and increased plasmatic absorption of the encapsulated GLP-1 analog (semaglutide). Our goal was to use our strategy to demonstrate a better outcome and a greater impact on the metabolic syndrome associated with MASLD and on liver disease progression with our strategy compared with the oral marketed version of semaglutide, Rybelsus®. Therefore, we studied the effect of our nanocarriers on a dietary mouse model of MASLD, the Western diet model, during a daily chronic treatment of 4 weeks. Overall, the results showed a positive impact of semaglutide-loaded lipid nanocapsules towards the normalization of glucose homeostasis and insulin resistance. In the liver, there were no significant changes in lipid accumulation, but an improvement in markers related to inflammation was observed. Overall, our strategy had a positive trend on the metabolic syndrome and at reducing inflammation, mitigating the progression of the disease. Oral administration of the nanosystem was more efficient at preventing the progression of the disease to more severe states when compared to the administration of Rybelsus®, as a suspension.


Subject(s)
Glucagon-Like Peptides , Lipids , Nanocapsules , Animals , Nanocapsules/administration & dosage , Glucagon-Like Peptides/administration & dosage , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/pharmacokinetics , Lipids/blood , Lipids/chemistry , Lipids/administration & dosage , Mice, Inbred C57BL , Male , Mice , Fatty Liver/drug therapy , Insulin Resistance , Liver/metabolism , Liver/drug effects , Disease Models, Animal
19.
J Control Release ; 369: 251-265, 2024 May.
Article in English | MEDLINE | ID: mdl-38493950

ABSTRACT

Immunotherapy is currently a standard of care in the treatment of many malignancies. However, predictable side effects caused by systemic administration of highly immunostimulatory molecules have been a serious concern within this field. Intratumoural expression or silencing of immunogenic and immunoinhibitory molecules using nucleic acid-based approaches such as plasmid DNA (pDNA) and small interfering RNA (siRNA), respectively, could represent a next generation of cancer immunotherapy. Here, we employed lipid nanoparticles (LNPs) to deliver either non-specific pDNA and siRNA, or constructs targeting two prominent immunotherapeutic targets OX40L and indoleamine 2,3-dioxygenase-1 (IDO), to tumours in vivo. In the B16F10 mouse model, intratumoural delivery of LNP-formulated non-specific pDNA and siRNA led to strong local immune activation and tumour growth inhibition even at low doses due to the pDNA immunogenic nature. Replacement of these non-specific constructs by pOX40L and siIDO resulted in more prominent immune activation as evidenced by increased immune cell infiltration in tumours and tumour-draining lymph nodes. Consistently, pOX40L alone or in combination with siIDO could prolong overall survival, resulting in complete tumour regression and the formation of immunological memory in tumour rechallenge models. Our results suggest that intratumoural administration of LNP-formulated pDNA and siRNA offers a promising approach for cancer immunotherapy.


Subject(s)
DNA , Immunotherapy , Mice, Inbred C57BL , Nanoparticles , Plasmids , RNA, Small Interfering , Animals , Immunotherapy/methods , RNA, Small Interfering/administration & dosage , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Plasmids/administration & dosage , DNA/administration & dosage , DNA/immunology , Mice , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Female , Cell Line, Tumor , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Lipids/chemistry , Lipids/administration & dosage , Drug Carriers/chemistry
20.
Curr Pharm Biotechnol ; 25(17): 2200-2217, 2024.
Article in English | MEDLINE | ID: mdl-38357950

ABSTRACT

Ovarian cancer poses a formidable health challenge for women globally, necessitating innovative therapeutic approaches. This review provides a succinct summary of the current research status on lipid-based nanocarriers in the context of ovarian cancer treatment. Lipid-based nanocarriers, including liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs), offer a promising solution for delivering anticancer drugs with enhanced therapeutic effectiveness and reduced adverse effects. Their versatility in transporting both hydrophobic and hydrophilic medications makes them well-suited for a diverse range of anticancer drugs. Active targeting techniques like ligand-conjugation and surface modifications have been used to reduce off-target effects and achieve tumour-specific medication delivery. The study explores formulation techniques and adjustments meant to enhance drug stability and encapsulation in these nanocarriers. Encouraging results from clinical trials and preclinical investigations underscore the promise of lipid-based nanocarriers in ovarian cancer treatment, providing optimism for improved patient outcomes. Notwithstanding these advancements, challenges related to clearance, long-term stability, and scalable manufacturing persist. Successfully translating lipidbased nanocarriers into clinical practice requires addressing these hurdles. To sum up, lipidbased nanocarriers are a viable strategy to improve the effectiveness of therapy for ovarian cancer. With their more focused medication administration and lower systemic toxicity, they may completely change the way ovarian cancer is treated and increase patient survival rates. Lipidbased nanocarriers need to be further researched and developed to become a therapeutically viable treatment for ovarian cancer.


Subject(s)
Antineoplastic Agents , Drug Carriers , Lipids , Nanoparticles , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/drug therapy , Lipids/chemistry , Lipids/administration & dosage , Drug Carriers/chemistry , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Animals
SELECTION OF CITATIONS
SEARCH DETAIL