Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
International Journal of Applied Pharmaceutics ; 15(3):1-11, 2023.
Article in English | EMBASE | ID: covidwho-20242785

ABSTRACT

Recent advancements in nanotechnology have resulted in improved medicine delivery to the target site. Nanosponges are three-dimensional drug delivery systems that are nanoscale in size and created by cross-linking polymers. The introduction of Nanosponges has been a significant step toward overcoming issues such as drug toxicity, low bioavailability, and predictable medication release. Using a new way of nanotechnology, nanosponges, which are porous with small sponges (below one microm) flowing throughout the body, have demonstrated excellent results in delivering drugs. As a result, they reach the target place, attach to the skin's surface, and slowly release the medicine. Nanosponges can be used to encapsulate a wide range of medicines, including both hydrophilic and lipophilic pharmaceuticals. The medication delivery method using nanosponges is one of the most promising fields in pharmacy. It can be used as a biocatalyst carrier for vaccines, antibodies, enzymes, and proteins to be released. The existing study enlightens on the preparation method, evaluation, and prospective application in a medication delivery system and also focuses on patents filed in the field of nanosponges.Copyright © 2023 The Authors.

2.
Cancer Research Conference ; 83(5 Supplement), 2022.
Article in English | EMBASE | ID: covidwho-2255725

ABSTRACT

During the COVID-19 pandemics we have all witnessed the clinical importance of mRNA as current vaccines and future therapeutics. mRNA therapies have a potential to revolutionize cancer treatment. Delivery of mRNA requires lipid nanoparticles (LNP) to protect the cargo from degradation. mRNA has a negative charge and depends on positively charged lipids to be encapsulated in LNP. These lipids can be either ionizable at certain pH or constantly cationic. Even though previous studies had evaluated the formulation properties of ionizable and cationic LNP systems, there is the need to understand their specificity in terms of mRNA delivery and protein expression in breast cancer tumor microenvironment. The objective of this work was to assess the kinetics of LNP cellular uptake and mRNA expression inv breast cancer (BC) cells and fibroblasts, the most frequent cell type in the tumor microenvironment cells, while studying the mechanisms involved in differential behaviors of LNP formulated with cationic and ionizable lipids. To achieve this goal mRNA-LNP containing ionizable lipids (LNP-A) and cationic lipids (LNP-B) were designed and formulated using Nanoassemblr Benchtop microfluidics mixer (Precision NanoSystems). mRNA-LNP were characterized for size, zeta potential using dynamic light scattering (DLS) and mRNA encapsulation efficiency using RiboGreen assay. LNP were tagged with rhodamine lipid to investigate the uptake kinetic and a reporter GFP mRNA to evaluate mRNA expression in murine 4T1 and human MCF7, MDA-231, SUM-159 and T47D breast cancer cells and BJ fibroblasts. Live fluorescence microscopy imaging, IncuCyte S3, was used to determine the LNP uptake and GFP mRNA expression. In vitro biocompatibility was assessed with WST-1 assay. Additionally, expression of mRNA delivered from LNP in tumor microenvironment was evaluated in vivo in a syngeneic 4T1 breast cancer model using mRNA luciferase and IVIS imaging. mRNA-LNPs possessed an average diameter of 77 - 107 nm, narrow size distribution, neutral zeta potential and high mRNA encapsulation efficiency (>94%). Our results demonstrated that mRNA expression was higher in breast cancer cells when delivered from LNP-A formulation and in BJ fibroblasts when delivered from LNP-B. LNP-A, the ionizable LNP, was tested in the breast cancer cells to confirm the efficacy of the delivery. The highest transfection efficacy, from high to low, T-47D, MCF7, SUM-159, 4T1 and MDA-231.We have further investigated the cellular uptake mechanisms of LNP using uptake pathway inhibitors for caveolae endocytosis, clathrin endocytosis, and phagocytosis. Our data confirm that there are differences in mechanisms that govern the uptake of mRNA LNP in breast cancer cells and fibroblasts. Clathrin-mediated endocytosis was active in 4T1 breast cancer cells for ionizable and cationic LNP. Interestingly, despite in vitro differences in uptake and mRNA expression, in vivo results show that both formulations efficiently delivered luciferasemRNA in the tumor microenvironment. Histology results demonstrated similar luciferase expression for both LNP in tumors. Additionally, we were able to confirm the prominent presence of fibroblast and similar distribution in the 4T1 subcutaneous model which could explain the similar efficacy of cationic and ionizable LNP. Understanding uptake and mRNA expression of different LNP formulations in the tumor microenvironment can help in achieving the necessary protein expression for breast cancer therapies. Furthermore, determining the most efficient carrier in early stages may reduce the time required for clinical translation. Acknowledgement: This research was supported in part by CPRIT Core for RNA Therapeutics and Research.

3.
Bioactive Materials ; 20:449-462, 2023.
Article in English | Scopus | ID: covidwho-2246587

ABSTRACT

The recent remarkable success and safety of mRNA lipid nanoparticle technology for producing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines has stimulated intensive efforts to expand nanoparticle strategies to treat various diseases. Numerous synthetic nanoparticles have been developed for pharmaceutical delivery and cancer treatment. However, only a limited number of nanotherapies have enter clinical trials or are clinically approved. Systemically administered nanotherapies are likely to be sequestered by host mononuclear phagocyte system (MPS), resulting in suboptimal pharmacokinetics and insufficient drug concentrations in tumors. Bioinspired drug-delivery formulations have emerged as an alternative approach to evade the MPS and show potential to improve drug therapeutic efficacy. Here we developed a biodegradable polymer-conjugated camptothecin prodrug encapsulated in the plasma membrane of lipopolysaccharide-stimulated macrophages. Polymer conjugation revived the parent camptothecin agent (e.g., 7-ethyl-10-hydroxy-camptothecin), enabling lipid nanoparticle encapsulation. Furthermore, macrophage membrane cloaking transformed the nonadhesive lipid nanoparticles into bioadhesive nanocamptothecin, increasing the cellular uptake and tumor-tropic effects of this biomimetic therapy. When tested in a preclinical murine model of breast cancer, macrophage-camouflaged nanocamptothecin exhibited a higher level of tumor accumulation than uncoated nanoparticles. Furthermore, intravenous administration of the therapy effectively suppressed tumor growth and the metastatic burden without causing systematic toxicity. Our study describes a combinatorial strategy that uses polymeric prodrug design and cell membrane cloaking to achieve therapeutics with high efficacy and low toxicity. This approach might also be generally applicable to formulate other therapeutic candidates that are not compatible or miscible with biomimetic delivery carriers. © 2022 The Authors

4.
Colloids and Surfaces B: Biointerfaces ; 220, 2022.
Article in English | EMBASE | ID: covidwho-2242220

ABSTRACT

Partitioning and effect of antiviral GC376, a potential SARS-CoV-2 inhibitor, on model lipid membranes was studied using dynamic light scattering (DLS), UV–VIS spectrometry, Excimer fluorescence, Differential scanning calorimetry (DSC) and Small- and Wide-angle X-ray scattering (SAXS/WAXS). Partition coefficient of GC376 between lipid and water phase was found to be low, reaching KP = 46.8 ± 18.2. Results suggest that GC376 partitions into lipid bilayers at the level of lipid head-groups, close to the polar/hydrophobic interface. Changes in structural and thermodynamic properties strongly depend on the GC376/lipid mole ratio. Already at lowest mole ratios GC376 induces increase of lateral pressures, mainly in the interfacial region of the bilayer. Hereby, the pre- and main-transition temperature of the lipid system increases, what is attributed to tighter packing of acyl chains induced by GC376. At GC376/DPPC ≥ 0.03 mol/mol we detected formation of domains with different GC376 content resulting in the lateral phase separation and changes in both, main transition temperature and enthalpy. The observed changes are attributed to the response of the system on the increased lateral stresses induced by partitioning of GC376. Obtained results are discussed in context of liposome-based drug delivery systems for GC376 and in context of indirect mechanism of virus replication inhibition.

5.
Indian Journal of Hematology and Blood Transfusion ; 38(Supplement 1):S125, 2022.
Article in English | EMBASE | ID: covidwho-2175096

ABSTRACT

Introduction: The COVID-19 pandemic has been a major public health concern throughout the world. Various ventures of vaccine candidates are being studied rigorously in this regard and one such candidate is the receptor binding domain (RBD) of spike protein which interacts with angiotensin converting enzyme 2 (ACE2) on the host cell's membrane. Exploiting this interaction, many scientists across the world attempted to develop mRNA vaccine against SARSCoV- 2. A major lacuna associated with mRNA vaccines is their delivery through a suitable carrier, especially given the stability issues associated with mRNA vaccines. Aims & Objectives: The aim of our research is to develop an efficient mode of delivery of the self-amplifying mRNA (saRNA) against COVID 19. We have developed small vesicles from erythrocyte ghosts, known as nanoerythrosomes, which are in the nanometre range and focussed on development of nanoerythrosomes for delivery of mRNA-based vaccines. Material(s) and Method(s): Nanoerythrosomes were prepared from erythrocytes using osmotic and ultrasonic frequency stress and loaded with saRNA vaccine candidate. Thereafter, the nanoerythrosomes were characterized using Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM) to confirm their homogeneity, integrity and size. The mRNA loaded nanoerythrosomes were used to deliver the mRNA in Vero E6 cells to evaluate its uptake. Result(s): The characterization of nanoerythrosomes using DLS and TEM revealed their size in the range of 100-200 nm. The delivery mediated by nanoerythrosomes was comparable to the Lipofectamine mediated uptake of saRNA indicating the excellent delivery efficacy of nanoerythrosomes. The added advantage of nanoerythrosomes mediated delivery is that they are rapidly taken up from blood by macrophages of the reticuloendothelial system (RES) that is present in liver, lung, and spleen. Thus the combination of saRNA and nanoerythrosomes can accelerate the uptake and antigen presentation in reticuloendothelial system and will provide an outstanding platform for the development of SARS-CoV2 vaccine. Conclusion(s): We developed a new approach to deliver mRNA vaccine candidates using nanoerythrosomes and successfully demonstrate its efficacy in vitro. This strategy can be further extended for the delivery of other vaccine candidates. (Figure Presented).

6.
European Journal of Hospital Pharmacy ; 29(SUPPL 1):A23-A24, 2022.
Article in English | EMBASE | ID: covidwho-1916404

ABSTRACT

Background and importance The importance of mRNA-based vaccines increased rapidly due to the COVID-19 pandemic. However, little is known on the challenges linked to handling shortages and extended stability of these new types of substance. Since vaccine remnants have to be discarded according to the Summary of Product Characteristics, we hypothesise that sterile filtration after pooling is suitable to save vaccine material for clinical application. Aim and objectives The aim of this pilot study was to compare quality parameters of remnants derived from ready-to-use mRNA vaccine solutions before and after sterile filtration. Therefore, we pooled mRNA vaccine solution remnants from Corminaty vials (BioNTech/Pfizer) and compared particle size, distribution and quantity of the lipoplexes. In addition, quantity and/or quality of the mRNA was determined. Material and methods Measurements of invisible particulates in the range 1-50 mm were performed by light obscuration according to the European Pharmacopoeia (10th edn). The size of lipoplexes was measured with nanoparticle tracking analysis (NTA) to determine hydrodynamic diameter and particle concentration. Dynamic light scattering was employed complementarily to the NTA technique to focus on particle size from 0.3 nm to 10 mm. The concentration, purity and integrity of the mRNA was analysed by ultraviolet (UV) spectrophotometry and capillary electrophoresis after mRNA purification. Results After pooling the remnants of the vials we found a substantial increase of particulates >1 mm when compared to fresh vaccine samples. This effect was likely due to contamination of the examined probes with particles from ambient air. As expected, all these particulates were eliminated by sterile filtration. Size distribution and concentration of the lipoplexes were comparable between unfiltered and filtered samples. With respect to the mRNA, we identified the fragment of interest in all examined samples. Sterile filtration did not change the concentration, purity and integrity of the mRNA. Conclusion and relevance Our results indicate that sterile filtration of mRNA-based vaccines eliminates particle contamination from the vaccine solution while the concentration of lipoplex nanoparticles was not altered. Moreover, neither the quantity nor quality of the mRNA was affected by the filtration process. The results of our pilot study provide the first data on the stability of mRNA vaccines and help to fill knowledge gap when dealing with these substances in hospital pharmacy.

7.
Topics in Antiviral Medicine ; 30(1 SUPPL):181-182, 2022.
Article in English | EMBASE | ID: covidwho-1880616

ABSTRACT

Background: SARS-CoV-2 is spread via airborne transmission. Mouthwashes containing virucidal compounds can help reduce viral spread. Here we show that cetylpyridinium chloride (CPC), a quaternary ammonium present in many oral mouthwashes, reduces SARS-CoV-2 infectivity by disrupting viral membranes both in vitro and in vivo. Methods: We tested the capacity of CPC-containing mouthwashes to inhibit SARS-CoV-2 entry into target cells by using a luciferase-based assay with a reporter lentivirus pseudotyped with the SARS-CoV-2 spike protein. The replication-competent SARS-CoV-2 B.1.1.7 and D614G variants were also assayed. Viral envelope disruption by CPC's virucidal effect was measured by dynamic light-scattering analyses (DSL). We confirmed these results by modifying an ELISA that detects the SARS-CoV-2 nucleocapsid (NC), which was used in the absence of its own lysis buffer. The effect of CPC in the saliva of individuals with CoVID-19 was assessed in a double-blind, placebo-controlled, randomized clinical trial. SARS-CoV-2 positive patients were randomized to gargle either water or 0.07% CPC mouthwash. The study outcomes were the SARS-CoV-2 log10 viral RNA load by RT-PCR and the NC protein levels by ELISA, both in saliva at 1h and 3h post-intervention. Results: CPC-containing mouthwashes inhibited SARS-CoV-2 viral fusion in vitro in a dose-dependent manner and decreased more than a 1000 times the viral TCID50 in target cells, regardless of the variant tested. The ELISA and the DSL analyses pointed to the effective disruption of the integrity of viral membranes after treatment with CPC. The clinical study performed with 105 patients showed no significant differences in viral RNA load at 1h and 3h post-treatment in saliva between placebo and CPC-treated groups. However, the levels of SARS-CoV-2 NC protein of lysed viruses were significantly higher in the CPC group at 1h and 3h post-intervention. Conclusion: CPC decreased more than a 1000 times the infectivity of SARS-CoV-2 in vitro and was effective against different SARS-CoV-2 variants. In CoVID-19 patients, the use of a 0.07% CPC mouthwash correlated with a statistically significant increase of NC protein levels in saliva, indicating enhanced disruption of viral particles. CPC-containing mouth rinses can represent a cost-effective measure to reduce SARS-CoV-2 infectivity in saliva, aiding to reduce viral transmission from infected individuals regardless of the variants they are infected with.

8.
Applied Sciences ; 12(2):720, 2022.
Article in English | ProQuest Central | ID: covidwho-1637221

ABSTRACT

Multi-layer graphene (2–10 layers), also called graphene nanoplatelets (GNPs), is a carbon-based nanomaterial (CBN) type with excellent properties desirable for many biomedical applications. Despite the promising advantages reported of GNPs, nanoscale materials may also present a potential hazard to humans. Therefore, in this study, the in vivo toxicity of these nanomaterials at a wide range of concentrations from 12.5 to 500 µg/mL was evaluated in the Caenorhabditis elegans model for 24 h (acute toxicity) and 72 h (chronic toxicity). Furthermore, their in vitro toxicity (from 0 to 10 µg/mL for 12 and 24 h), proliferative activity at 72 and 96 h, and their effect on the expression of thirteen genes in human keratinocytes HaCaT cells were studied. The physico-chemical and morphological aspects of the GNPs used in this study were analyzed by Raman scattering spectroscopy, electron microscopy, zeta potential as a function of pH, and particle size measurements by dynamic light scattering. The results of this study showed that GNPs showed in vivo non-toxic concentrations of 25 and 12.5 µg/mL for 24 h, and at 12.5 µg/mL for 72 h. Moreover, GNPs present time-dependent cytotoxicity (EC50 of 1.142 µg/mL and 0.760 µg/mL at 12 h and 24 h, respectively) and significant proliferative activity at the non-toxic concentrations of 0.005 and 0.01 μg/mL in the HaCaT cell line. The gene expression study showed that this multi-layer-graphene is capable of up-regulating six of the thirteen genes of human keratinocytes (SOD1, CAT, TGFB1, FN1, CDH1, and FBN), two more genes than other CBNs in their oxidized form such as multi-layer graphene oxide. Therefore, all these results reinforce the promising use of these CBNs in biomedical fields such as wound healing and skin tissue engineering.

9.
Blood ; 138:292, 2021.
Article in English | EMBASE | ID: covidwho-1577299

ABSTRACT

VITT is an immune-based complication of adenoviral-based vaccines used to immunize against SARS_CoV2. The antibodies in VITT have been described as directed at the platelet-specific chemokine PF4 (CXCL4). While the clinical course and target chemokine in VITT has much in common with the better-known thrombocytopenic/prothrombotic disorder, heparin-induced thrombocytopenia (HIT), which involves antibodies directed against PF4 bound to the polyanion heparin, the specific loci where VITT and PF4/polyanion HIT antibodies bind appear to differ in studies using alanine-scanning mutations of PF4 (Nature, 2021. DOI: 10.1038/s41586-021-03744-4). The VITT antigenic site localizes to a heparin-binding domain. Unlike the dominant HIT locus, the VITT locus is conserved not only between human and mouse PF4, but also between PF4 and the related platelet-specific chemokine NAP2 (CXCL7). NAP2 is also expressed and stored in platelet alpha-granules and is present in equimolar concentrations to PF4. Unlike PF4, NAP2 avidly binds the chemokine receptor CXCR2 and strongly activates neutrophils. We now show that antibodies from patients who developed VITT after both AstraZeneca (AZ) or Johnson and Johnson (JJ) adenoviral vaccines, unlike HIT antibodies, recognize mouse PF4 (Figure 1A). More importantly, both AZ and JJ VITT antibodies bound NAP2, while none of the HIT antibodies tested bound PF4 or NAP2 in the absence of heparin (Figure 1A). These results are consistent with the alanine-scanning studies that distinguish the HIT and VITT binding sites. Dynamic light scattering (DLS) showed that NAP2 and PF4 bind to the adenoviral vectors, including Ad5 and the AZ vector ChAdOx5, which leads to expression of SARS_CoV2 spike protein. ChAdOx2 vaccine and CsCl 2-purified ChAdOx2 bound to both proteins, but form larger complexes with NAP2 than with PF4 even at lower concentrations of this chemokine (Figure 1C). Removal of anti-PF4 antibodies by hPF4-Sepharose abrogated PF4-dependent binding, but did not significantly reduce binding to NAP2 (not shown), indicating that VITT plasma contains discrete pools of anti-PF4 and anti-NAP2 antibodies that may have distinct functional properties. Sandwich ELISA (not shown) and Western blot analysis of purified VITT IgG demonstrates the presence of hPF4-IgG and NAP2-IgG immune complexes in purified patient's IgG (Figure 2A). Functional studies show that both PF4 and NAP2 can activate platelets in the presence of VITT antibodies. Anti-PF4-depleted VITT IgG fraction retains the ability to activate platelets in the presence of NAP2 (Figure 2B). Thus, unlike HIT, VITT appears to target a shared antigenic site on the related chemokines PF4 and NAP2. This raises the question as to whether NAP2, as one the most abundant platelet chemokines released from activated platelets, is involved in the initiation and propagation of the immunothrombotic response. Additional studies are needed to see whether NAP2, which can potently and specifically activate neutrophils via CXCLR2, contributes to the specific thromboinflammatory phenotype seen in VITT. We propose using FcgammaRIIA+ mice that concurrently express human PF4 and NAP2 and specific knockout of each chemokine, available in our group, to further understand the pathogenesis of VITT and its thrombocytopenic/ prothrombotic phenotype. [Formula presented] Disclosures: Padmanabhan: Veralox Therapeutics: Membership on an entity's Board of Directors or advisory committees. Cines: Dova: Consultancy;Rigel: Consultancy;Treeline: Consultancy;Arch Oncol: Consultancy;Jannsen: Consultancy;Taventa: Consultancy;Principia: Other: Data Safety Monitoring Board.

SELECTION OF CITATIONS
SEARCH DETAIL