Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 271(Pt 2): 132426, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38820904

ABSTRACT

Eye-related diseases, specifically retinal dystrophy (RD) conditions, are the leading cause of blindness worldwide. Gene addition, regulation, or editing could potentially treat such diseases through gene expression regulation. CRISPR/Cas9 gene editing is one of the most prominent and precise gene editing tools which could be employed to edit genes related to the dystrophic condition. However, CRISPR/Cas9 faces in vivo delivery challenges due to its high molecular weight, negative charge, prone to degradation in the presence of nucleases and proteases, poor cellular degradation, etc., which makes it challenging to adopt for therapeutic applications. We developed cRGD-modified lipopolymeric nanoplexes loaded with Cas9 RNPs with a particle size and zeta potential of 175 ±â€¯20 nm and 2.15 ±â€¯0.9 mV, respectively. The cRGD-modified lipopolymeric nanoplexes were stable for 194 h and able to transfect >70 % ARPE-19 and NIH3T3 cells with an Indel frequency of ~40 % for the VEGF-A gene. The cRGD-modified lipopolymeric nanoplexes found good vitreous mobility and could transfection retinal cells in vivo after 48 h of intravitreal injection in Wistar Rats. Moreover, in vivo VEGFA gene editing was ~10 % with minimal toxicities. Collectively, the cRGD-modified lipopolymeric nanoplexes were found to have extreme potential in delivering CRISPR/Cas9 RNPs payload to the retinal tissues for therapeutic applications.


Subject(s)
Gene Editing , Animals , Gene Editing/methods , Mice , Rats , Humans , NIH 3T3 Cells , CRISPR-Cas Systems , Oligopeptides/chemistry , Rats, Wistar , Transfection/methods , Vascular Endothelial Growth Factor A/genetics , Peptides, Cyclic
3.
Eur J Pharm Biopharm ; 195: 114175, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38185191

ABSTRACT

Exosomes, biogenic nano-vesicles, are renowned for their ability to encapsulate diverse payloads, however the systematic development and validation of exosomal formulation with significant biological implications have been overlooked. Herein, we developed and validated Exo-DTX, a QbD-driven optimized RAW 264.7 cell derived exosomal anti-cancer formulation of docetaxel (DTX) and evaluate its anti-metastatic and apoptotic efficacy in TNBC 4T1 cells. RAW264.7-derived exosomes were having particle size (112.5 ± 21.48 nm) and zeta-potential (-10.268 ± 3.66 mV) with polydispersity (PDI:0.256 ± 0.03). The statistical optimization of exosomes (200 µg) with Exo: DTX ratio 4:1 confirmed encapsulation of 23.60 ± 1.54 ng DTX/ µg exosomes. Exo-DTX (∼189 nm, -11.03 mV) with 100 ng/ml DTX as payload exhibited ∼5 folds' improvement in IC50 of DTX and distinct cytoskeletal deformation in TNBC 4T1 cells. It also has shown enormous Filamentous actin (F-actin) degradation and triggered apoptosis explained Exo-DTX's effective anti-migratory impact with just 2.6 ± 6.33 % wound closure and 4.56 ± 1.38 % invasion. The western blot confirmed that Exo-DTX downregulated migratory protein EGFR and ß1-integrin but raised cleaved caspase 3/caspase 3 (CC3/C3) ratio and BAX/BCL-2 ratio by about 2.70 and 4.04 folds respectively. The naive RAW 264.7 exosomes also contributed positively towards the effect of Exo-DTX formulation by suppressing ß1-integrin expression and increasing the CC3/C3 ratio in TNBC 4T1 cells as well. Additionally, significant improvement in PK parameters of Exo-DTX was observed in comparison to Taxotere, 6-folds and 3.04-folds improved t1/2 and Vd, proving the translational value of Exo-DTX formulation. Thus, the Exo-DTX so formulated proved beneficial in controlling the aggressiveness of TNBC wherein, naive exosomes also demonstrated beneficial synergistic anti-proliferative effect in 4T1.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Docetaxel/pharmacokinetics , Caspase 3 , Macrophages , Integrins , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor
4.
J Pharmacol Exp Ther ; 388(1): 81-90, 2024 01 02.
Article in English | MEDLINE | ID: mdl-37863489

ABSTRACT

Dysregulation of various glucoregulatory hormones lead to failure of insulin monotherapy in patients with diabetes mellitus due to various reasons, including severe hypoglycemia, glycemic hypervariability, and an increased risk of microvascular complications. However, pramlintide as an adjunct to insulin therapy enhances glucagon suppression and thereby offers improved glycemic control. Clinical studies have shown that pramlintide improves glycemic control, reduces postprandial glucose excursions, and promotes weight loss in patients with type 1 and type 2 diabetes. Although clinical benefits of pramlintide are well reported, there still exists a high patient resistance for the therapy, as separate injections for pramlintide and insulin must be administered. Although marketed insulin formulations generally demonstrate a peak action in 60-90 minutes, pramlintide elicits its peak concentration at around 20-30 minutes after administration. Thus, owing to the significant differences in pharmacokinetics of exogenously administered pramlintide and insulin, the therapy fails to elicit its action otherwise produced by the endogenous hormones. Hence, strategies such as delaying the release of pramlintide by using inorganic polymers like silica, synthetic polymers like polycaprolactone, and lipids have been employed. Also, approaches like noncovalent conjugation, polyelectrolyte complexation, and use of amphiphilic excipients for codelivery of insulin and pramlintide have been explored to address the issues with pramlintide delivery and improve patient adherence to the therapy. This approach may usher in a new era of diabetes management, offering patients multiple options to tailor their treatment and improve their quality of life. SIGNIFICANCE STATEMENT: To our knowledge, this is the first report that summarizes various challenges in insulin and pramlintide codelivery and strategies to overcome them. The paper also provides deeper insights into various novel formulation strategies for pramlintide that could further broaden the reader's understanding of peptide codelivery.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Humans , Insulin , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Quality of Life , Polymers , Blood Glucose
5.
J Mater Chem B ; 11(45): 10859-10872, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37938124

ABSTRACT

Despite various treatment modalities for breast cancer, it still persists as one of the most diagnosed types of cancer in females. The recent investigations in the epigenetics of breast cancer reveal several aberrations in the expression levels of various HDAC enzymes. Henceforth, the present work entails the formulation and characterization of a lipid polymer-based hybrid nanoparticulate (LPN) system for delivery of an epigenetic modulator drug, Belinostat, for its clinical application in breast cancer. The size of Belinostat nanoparticles prepared using a modified hot homogenization method was found to be 166.6 ± 19.95 nm with an encapsulation efficiency of 94.5 ± 5.1%. In vitro characterization for cytotoxicity, cellular uptake, and protein expression in two different breast cancer cells, 4T1 and MCF 7, revealed the superiority of the formulation in comparison with the free drug in MCF 7 cells. Subsequently, the behaviour of the formulation in in vivo settings of healthy and breast cancer xenograft bearing animals was analyzed using pharmacokinetic and biodistribution studies. The results revealed that the formulation demonstrated multi-fold improvement in the pharmacokinetic parameters in tumor bearing animals when compared with the free drug while no difference in pharmacokinetic behaviour was observed in healthy animals indicating the altered biodistribution and specificity of the formulation in breast tumor. This was confirmed by the biodistribution studies exhibiting 20-fold improved uptake and retention of the nanoparticulate formulation in tumor tissues of the animal model at the end of 4 h. Thus, the developed LPN system holds potential to act as a novel drug delivery system for Belinostat with several advantages over the free drug.


Subject(s)
Breast Neoplasms , Animals , Female , Humans , Breast Neoplasms/drug therapy , Polymers/metabolism , Tissue Distribution , Cell Line, Tumor , Lipids
6.
J Control Release ; 359: 161-174, 2023 07.
Article in English | MEDLINE | ID: mdl-37182806

ABSTRACT

Glioblastoma multiforme (GBM) is the deadliest brain tumor with a poor prognosis and limited therapeutic options. Temozolomide (TMZ) is the first-line chemotherapeutic agent used for the treatment of GBM; however, it suffers from several limitations, including short half-life, rapid metabolism, <1% brain bioavailability, methyl guanine methyl transferase (MGMT) based chemoresistance, and hematological toxicities. Several approaches have been adopted to overcome these limitations, particularly by using nanotechnology-based systems, but its physicochemical properties make TMZ challenging to load into these nanocarriers. In the current research, we conjugated TMZ with different fatty acids, i.e., linoleic acid (LA), oleic acid (OA), and palmitic acid (PA), to obtain TMZ-fatty acid conjugates, which are comparatively hydrophobic, less prone to degradation and potent. These conjugates were thoroughly characterized using 1H NMR spectroscopy, high-resolution mass spectrometry (HR-MS), and reverse phase-high performance liquid chromatography (RP-HPLC). The synthesized conjugates, namely Temozolomide-oleic acid (TOA,6R1), Temozolomide-linoleic acid (TLA, 6R2), and Temozolomide-palmitic acid (TPA, 6R3), showed an IC50 of 101.4, 67.97, and 672.04 µM, respectively in C6 cells and 428.257, 366.43 and 413.69 µM, respectively in U87-MG cells. On the other hand, the free TMZ showed an IC50 of >1000 µM and 564.23 µM in C6 and U87-MG, respectively. Further, the in vivo efficacy of the TMZ-fatty acid conjugates was evaluated in the C6-induced orthotropic rat glioblastoma model, wherein the TMZ-fatty acid conjugate showed improved survival rate (1.6 folds) and overall health of the animals. Collectively, the conjugation of fatty acids with TMZ improves its anticancer potential against glioblastoma multiforme (GBM).


Subject(s)
Brain Neoplasms , Glioblastoma , Rats , Animals , Temozolomide/therapeutic use , Glioblastoma/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Alkylating/pharmacology , Fatty Acids , Cell Line, Tumor , Brain Neoplasms/metabolism , Linoleic Acids/therapeutic use , Palmitic Acids/therapeutic use , Oleic Acids/therapeutic use , Drug Resistance, Neoplasm , Xenograft Model Antitumor Assays
7.
Macromol Rapid Commun ; 44(14): e2300101, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37186473

ABSTRACT

CRISPR/Cas9 has proven its accuracy and precision for gene editing by making a double-strand break at the predetermined site. Despite being a mainstream gene editing tool, CRISPR/Cas9 has limitations for its in vivo delivery due to the physico-chemical properties such as high molecular weight, supranegative charge, degradation in the presence of nucleases, etc. Hereby, a cationic lipopolymer is explored for its efficiency in delivering CRISPR/Cas9 plasmid (pCas9) in vitro and in vivo. The lipopolymer is utilized to form blank cationic nanoplexes having a zeta potential of +15.8 ± 0.7 mV. Being cationic, the blank nanoplexes are able to condense the pCas9 plasmid at a ratio of 1:20 with a complexation efficiency of ≈98% and show a size and zeta potential of ≈141 ± 16 nm and 4.2 mV ± 0.7, respectively. The pCas9-loaded nanoplexes show a transfection efficiency of ≈69% in ARPE-19 cells and show ≈22% of indel frequency, indicating the successful translation of Cas9 protein and guide RNA in the cytosol. Further, they are found to be stable under in vivo environment when given intravenously in Swiss albino mice. These lipopolymeric nanoplexes can be a potential carrier for CRISPR plasmids for genome editing applications.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , Mice , CRISPR-Associated Protein 9/metabolism , Transfection , Plasmids/genetics
8.
Life Sci ; 322: 121621, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37001803

ABSTRACT

AIM: Exosomes, as a nanocarrier for the co-delivery of biologicals and small anticancer molecules is yet in its infancy. Herein, we investigated hUCBMSC derived exosomes as a biogenic nanocarrier for the co-delivery of tumor suppressor miR-125a and microtubule destabilizing Docetaxel (DTX) to target the proliferative and migratory aggressiveness of the murine TNBC 4T1 cells. MAIN METHODS: In this study, hUCBMSCs from the human umbilical cord blood cells (hUCB) were successfully transfected with miR-125a. Thereafter, DTX was encapsulated into both non-transfected and transfected exosomes by optimized mild sonication-incubation technique. The anticancer efficiency of hUCBMSC Exo-DTX and miR-125a Exo-DTX was compared by MTT and morphometric assay. The prominent anti-metastatic behaviour of the latter was confirmed by in-vitro wound healing and transwell invasion assay. Further, the synergistic effect of miR-125a and DTX was confirmed by F-actin and nuclear degradation by confocal and FESEM assay. KEY FINDINGS: hUCBMSC exosomes exhibited DTX payload of 8.86 ± 1.97 ng DTX/ µg exosomes and miRNA retention capacity equivalent to 12.31 ± 5.73 %. The co-loaded formulation (miR-125a Exo-DTX) exhibited IC50 at 192.8 ng/ml in 4T1 cells, which is almost 2.36 folds' lower than the free DTX IC50 (472.8 ng/ml). Additionally, miR-125a Exo-DTX treatment caused wound broadening upto 6.14±0.38 % while treatment with free DTX and miR-125a exosomes alone caused 18.71±4.5 % and 77.36±10.4 % of wound closure respectively in 36 h. miR-125a Exo-DTX treatment further exhibited significantly reduced invasiveness of 4T1 cells (by 3.5 ± 1.8 %) along with prominent cytoskeletal degradation and nuclear deformation as compared to the miR-125a exosomes treated group. The miR-125a expressing DTX loaded exosomal formulation clearly demonstrated the synergistic apoptotic and anti-migratory efficiency of the miR-125a Exo-DTX. SIGNIFICANCE: The synergistic anticancer and anti-metastatic effect of miR-125a Exo-DTX was observed due to presence of both DTX and miR-125a as the cargo of hUCBMSC derived exosomes.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Humans , Animals , Mice , Docetaxel/pharmacology , Exosomes/metabolism , Fetal Blood/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism
9.
Int J Pharm ; 631: 122508, 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36539166

ABSTRACT

Asiaticoside (AST) is a naturally available phytoconstituent that enables effective wound healing mainly by promoting collagen biosynthesis. However, the physicochemical nature of AST such as high molecular weight (959.12 g/mol), poor water solubility and poor permeability limits its therapeutic effects. This study aims to develop Asiaticoside polymeric nanoparticles (AST PNP) embedded in a gelatin based biodegradable hydrogel (15 % w/v) for application in the wound cavity to enable sustained release of AST and enhance its therapeutic effects. The AST PNP were fabricated in the desired size range (168.4 nm; PDI (0.09)) and the morphology, rate of fluid uptake, rate of water loss, and water vapor transmission rate of AST PNP incorporated hydrogel were determined. AST PNP gel showed porous structural morphology and possessed ideal characteristics as a graft for wound healing. The drug release kinetics and cellular uptake of AST PNP were investigated wherein, AST PNP demonstrated sustained release profile upto 24 h in comparison to free AST (complete release within 6 h) and exhibited an enhanced intra-cellular uptake in fibroblasts within 3 h compared to the free drug. In-vitrocell culture studies also demonstrated significant proliferation and migration of fibroblasts in the presence of AST PNP. Additionally, AST PNP gel upon application to the wounds of diabetic rats depicted improved wound healing efficacy in terms of improved collagen biosynthesis, upregulated COL-1 protein level (∼1.85 fold vs free AST), and enhanced expression of α-SMA compared to control groups. Altogether, formulation of AST as polymeric nanoparticles in a gel based carrier offered significant improvement in the therapeutic properties of AST for the management of diabetic wounds.


Subject(s)
Diabetes Mellitus, Experimental , Nanoparticles , Rats , Animals , Diabetes Mellitus, Experimental/drug therapy , Delayed-Action Preparations/pharmacology , Wound Healing , Collagen/chemistry , Nanoparticles/chemistry , Hydrogels/chemistry
10.
ACS Biomater Sci Eng ; 9(1): 20-39, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36524837

ABSTRACT

"Cancer" is a dreadful immune-pathological condition that is characterized by anti-inflammatory and tumorigenic responses, elicited by the infiltrating immune cells in the vicinity of an uncontrollably proliferative tumor in the tumor microenvironment (TME). The TME offers a conducive microenvironment that supports cancer cell survival by modulating the host immune defense. Recent advancement in exosomal research has shown exosomes, originating from immune cells as well as the cancer cells, have immense potential for suppressing cancer progression and survival in the TME. Additionally, exosomes, irrespective of their diverse sources, have been reported to be efficient nanocarriers for cancer therapeutics with the ability for targeted delivery due to their biogenic nature, ease of cellular uptake, and scope for functionalization with biomolecules like peptides, aptamers, targeting ligands, etc. Immune cell-derived exosomes per se have been found efficacious against cancer owing to their immune-stimulant properties (in either naive or antigen primed form) even without loading any of cancer therapeutics or targeting ligand conjugation. Nevertheless, exosomes are being primarily explored as nanovesicular carriers for therapeutic molecules with different loading and targeting strategies, and the synergism between immunotherapeutic behavior of exosomes and the anticancer effect of the therapeutic molecules is yet to be explored. Hence, this review focuses specifically on the possible strategies to modulate the immunological nature of the source immune cells to obtain immune stimulant exosomes and bring these into the spotlight as chemo-immunotherapeutic nanovesicles, that can easily target and modulate the TME.


Subject(s)
Exosomes , Neoplasms , Humans , Exosomes/pathology , Exosomes/physiology , Tumor Microenvironment , Neoplasms/drug therapy , Immunotherapy
11.
Asian J Pharm Sci ; 17(2): 153-176, 2022 Mar.
Article in English | MEDLINE | ID: mdl-36320315

ABSTRACT

CRISPR/Cas, an adaptive immune system in bacteria, has been adopted as an efficient and precise tool for site-specific gene editing with potential therapeutic opportunities. It has been explored for a variety of applications, including gene modulation, epigenome editing, diagnosis, mRNA editing, etc. It has found applications in retinal dystrophic conditions including progressive cone and cone-rod dystrophies, congenital stationary night blindness, X-linked juvenile retinoschisis, retinitis pigmentosa, age-related macular degeneration, leber's congenital amaurosis, etc. Most of the therapies for retinal dystrophic conditions work by regressing symptoms instead of reversing the gene mutations. CRISPR/Cas9 through indel could impart beneficial effects in the reversal of gene mutations in dystrophic conditions. Recent research has also consolidated on the approaches of using CRISPR systems for retinal dystrophies but their delivery to the posterior part of the eye is a major concern due to high molecular weight, negative charge, and in vivo stability of CRISPR components. Recently, non-viral vectors have gained interest due to their potential in tissue-specific nucleic acid (miRNA/siRNA/CRISPR) delivery. This review highlights the opportunities of retinal dystrophies management using CRISPR/Cas nanomedicine.

12.
J Control Release ; 350: 494-513, 2022 10.
Article in English | MEDLINE | ID: mdl-35985493

ABSTRACT

Temozolomide (TMZ), an imidazotetrazine, is a second-generation DNA alkylating agent used as a first-line treatment of glioblastoma multiforme (GBM). It was approved by FDA in 2005 and declared a blockbuster drug in 2008. Although TMZ has shown 100% oral bioavailability and crosses the blood-brain barrier effectively, however it suffers from limitations such as a short half-life (∼1.8 h), rapid metabolism, and lesser accumulation in the brain (∼10-20%). Additionally, development of chemoresistance has been associated with its use. Since it is a potential chemotherapeutic agent with an unmet medical need, advanced delivery strategies have been explored to overcome the associated limitations of TMZ. Nanocarriers including liposomes, solid lipid nanoparticles (SLNs), nanostructure lipid carriers (NLCs), and polymeric nanoparticles have demonstrated their ability to improve its circulation time, stability, tissue-specific accumulation, sustained release, and cellular uptake. Because of the appreciable water solubility of TMZ (∼5 mg/mL), the physical loading of TMZ in these nanocarriers is always challenging. Alternatively, the conjugation approach, wherein TMZ has been conjugated to polymers or small molecules, has been explored with improved outcomes in vitro and in vivo. This review emphasized the practical evidence of the conjugation strategy to improve the therapeutic potential of TMZ in the treatment of glioblastoma multiforme.


Subject(s)
Brain Neoplasms , Glioblastoma , Alkylating Agents/therapeutic use , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Cell Line, Tumor , Delayed-Action Preparations/therapeutic use , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Humans , Lipids/chemistry , Liposomes/therapeutic use , Nanoparticles , Polymers/therapeutic use , Temozolomide/therapeutic use , Water
13.
J Mater Chem B ; 10(37): 7634-7649, 2022 09 28.
Article in English | MEDLINE | ID: mdl-35946380

ABSTRACT

sgRNA/Cas9 ribonucleoproteins (RNPs) provide a site-specific robust gene-editing approach avoiding the mutagenesis and unwanted off-target effects. However, the high molecular weight (∼165 kDa), hydrophilicity and net supranegative charge (∼-20 mV) hinder the intracellular delivery of these RNPs. In the present study, we have prepared cationic RNPs lipopolymeric nanoplexes that showed a size of 117.3 ± 7.64 nm with +6.17 ± 1.04 mV zeta potential and >90% entrapment efficiency of RNPs. Further, these RNPs lipopolymeric nanoplexes showed good complexation efficiency and were found to be stable for 12 h with fetal bovine serum. These RNPs lipopolymeric nanoplexes did not induce any significant cytotoxicity in HEK293T cells, and were efficiently uptaken via a clathrin-mediated pathway with optimal transfection efficiency and nuclear localization after 48 h. Further, HEK293T cells having the mGFP insert were used as a cell line model for gene editing, wherein the loss of the mGFP signal was observed as a function of gene editing after transfection with mGFP targeting RNPs lipopolymeric nanoplexes. Further, the T7 endonuclease and TIDE assay data showed a decent gene editing efficiency. Additionally, the lipopolymeric nanoplexes were able to transfect muscle cells in vivo, when injected intra-muscularly. Collectively, this study explored the potential of cationic lipopolymeric nanoplexes for delivering gene-editing endonucleases.


Subject(s)
CRISPR-Cas Systems , Ribonucleoproteins , CRISPR-Cas Systems/genetics , Clathrin/genetics , Clathrin/metabolism , Endonucleases/genetics , Endonucleases/metabolism , HEK293 Cells , Humans , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Serum Albumin, Bovine/metabolism
14.
Mol Pharm ; 19(7): 1977-1998, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35481377

ABSTRACT

Diabetes mellitus is a chronic manifestation characterized by high levels of glucose in the blood resulting in several complications including diabetic wounds and ulcers, which predominantly require a longer duration of treatment and adversely affect the quality of life of the patients. Nanotechnology-based therapeutics (both intrinsic and extrinsic types) have emerged as a promising treatment in diabetic foot ulcer/chronic wounds owing to their unique characteristics and specific functional properties. In this review, we have focused on the significance of the use of lipids in the healing of diabetic ulcers, their interaction with the injured skin, and recent trends in lipid-based nanocarriers for the healing of diabetic wounds. Lipid nanocarriers are also being investigated for gene therapy in diabetic wound healing to encapsulate nucleic acids such as siRNA and miRNA, which could silence the expression of inflammatory cytokines overexpressed in chronic wounds. Additionally, these are also being explored for encapsulating proteins, peptides, growth factors, and other biological genetic material as therapeutic agents. Lipid-based nanocarriers encompassing a wide variety of carriers such as liposomes, niosomes, ethosomes, solid lipid nanoparticles, and lipidoid nanoparticles that are explored for the treatment of foot ulcers supplemented with relevant research studies have been discussed in the present review. Lipid-based nanodrug delivery systems have demonstrated promising wound healing potential, particularly in diabetic conditions due to the enhanced efficacy of the entrapped active molecules.


Subject(s)
Diabetes Mellitus , Diabetic Foot , Diabetic Foot/drug therapy , Diabetic Foot/metabolism , Drug Delivery Systems , Humans , Lipids , Liposomes , Nanoparticles , Quality of Life
15.
J Med Chem ; 64(19): 14217-14229, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34581574

ABSTRACT

Several drug-fatty acid (FA) prodrugs have been reported to exhibit desirable physicochemical and pharmacological profile; however, comparative beneficial effects rendered by different FAs have not been explored. In the present study, four different FAs (linoleic acid, oleic acid, palmitic acid, and α-lipoic acid) were selected based on their chain length and degree of unsaturation and conjugated to Lisofylline (LSF), an antidiabetic molecule to obtain different drug-FA prodrugs and characterized for molecular weight, hydrophobicity, purity, self-assembly, and efficacy in vitro and in vivo in type 1 diabetes model. Prodrugs demonstrated a 2- to 6-fold increase in the plasma half-life of LSF. Diabetic animals treated with prodrugs, once daily for 5 weeks, maintained a steady fasting blood glucose level with a significant increase in insulin level, considerable restoration of biochemical parameters, and preserved ß-cells integrity. Among the different LSF-FA prodrugs, LSF-OA and LSF-PA demonstrated the most favorable physicochemical, systemic pharmacokinetic, and pharmacodynamic profiles.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Fatty Acids/pharmacology , Hypoglycemic Agents/pharmacology , Prodrugs/pharmacology , Animals , Cell Line , Diabetes Mellitus, Type 1/chemically induced , Dose-Response Relationship, Drug , Fatty Acids/chemistry , Hypoglycemic Agents/chemistry , Mice , Molecular Structure , Prodrugs/chemistry , Rats , Rats, Wistar , Streptozocin , Structure-Activity Relationship
16.
AAPS PharmSciTech ; 22(7): 238, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34561775

ABSTRACT

Lipid-polymer hybrid nanoparticles display several benefits over either lipid and/or polymer based systems with respect to enhanced drug loading, good colloidal stability, sustained release profile, and high cellular uptake. The present work rivets on development and evaluation of vitamin D3-loaded monolithic lipid-polymer hybrid nanoparticles (VD3/LPHNPs) for their in vivo anti-psoriatic efficacy. These LPHNPs were prepared using a hot homogenization method and exhibited spherical morphology with a lower particle size (123.1 nm) with narrow PDI (0.234) and efficient encapsulation (76.80%). Further, these LPHNPs demonstrated a sustained release profile of VD3 for up to 3 days following a Korsemeyer-Peppas release model. Further, VD3/LPHNPs were formulated into a topical gel containing 0.005% w/w of VD3. Rheological data suggested that the product exhibited non-newtonian flow properties with characteristic shear-thinning and variable thixotropy features that are desirable for topical formulation. The successful formation of gel structure and its long-term stability were confirmed from the oscillatory studies such as amplitude and frequency sweep tests. In vivo efficacy assessment in imiquimod-induced psoriatic mouse model demonstrated enhanced anti-psoriatic activity of VD3 with improved PASI score when delivered as LPHNPs gel as compared to the free VD3 gel that were further supported by histopathology and immunohistochemistry.


Subject(s)
Cholecalciferol , Imiquimod , Nanoparticles , Psoriasis , Animals , Mice , Particle Size , Psoriasis/drug therapy
17.
Mater Sci Eng C Mater Biol Appl ; 128: 112305, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34474856

ABSTRACT

In spite of established evidence of the synergistic combination of hydrophobic anticancer molecule and microRNA for breast cancer treatment, their in vivo delivery has not been realized owing to their instability in the biological milieu and varied physicochemical properties. The present work reports folate targeted hybrid lipo-polymeric nanoplexes for co-delivering DTX and miR-34a. These nanoplexes exhibited a mean size of 129.3 nm with complexation efficiency at an 8:1 N/P ratio. The obtained nanoplexes demonstrated higher entrapment efficiency of DTX (94.8%) with a sustained release profile up to 85% till 48 h. Further, an improved transfection efficiency in MDA-MB-231 and 4T1 breast cancer cells was observed with uptake primarily through lipid-raft and clathrin-mediated endocytosis. Further, nanoplexes showed improved cytotoxicity (~3.5-5 folds), apoptosis (~1.6-2.0 folds), and change in expression of apoptotic genes (~4-7 folds) compared to the free treatment group in breast cancer cells. In vivo systemic administration of FA-functionalized DTX and FAM-siRNA-loaded nanoplexes showed an improved area under the curve (AUC) as well as circulation half-life compared to free DTX and naked FAM-labelled siRNA. Acute toxicity studies of the cationic polymer showed no toxicity at a dose equivalent to 10 mg/kg based on the hematological, biochemical, and histopathological examination.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , MicroRNAs/administration & dosage , Nanoparticles , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Docetaxel/pharmacology , Drug Carriers/therapeutic use , Female , Folic Acid , Humans , MicroRNAs/genetics , Polymers/therapeutic use
18.
Biomaterials ; 275: 120953, 2021 08.
Article in English | MEDLINE | ID: mdl-34218051

ABSTRACT

Designing grafted biodegradable polymers with tailored multi-functional properties is one of the most researched fields with extensive biomedical applications. Among many biodegradable polymers, polycarbonates have gained much attention due to their ease of synthesis, high drug loading, and excellent biocompatibility profiles. Among various monomers, 2,2-bis(hydroxymethyl) propionic acid (bis-MPA) derived cyclic carbonate monomers have been extensively explored in terms of their synthesis as well as their polymerization. Since the late 90s, significant advancements have been made in the design of bis-MPA derived cyclic carbonate monomers as well as in their reaction schemes. Currently, bis-MPA derived polycarbonates have taken a form of an entire platform with a multitude of applications, the latest being in the field of nanotechnology, targeted drug, and nucleic acid delivery. The present review outlines an up to date developments that have taken place in the last two decades in the design, synthesis, and biomedical applications of bis-MPA derived cyclic carbonates and their (co)polymers.


Subject(s)
Pharmaceutical Preparations , Polymers , Carbonates , Polycarboxylate Cement , Propionates
19.
Acta Pharm Sin B ; 11(4): 903-924, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33996406

ABSTRACT

The beneficial or deleterious effects of nanomedicines emerge from their complex interactions with intracellular pathways and their subcellular fate. Moreover, the dynamic nature of plasma membrane accounts for the movement of these nanocarriers within the cell towards different organelles thereby not only influencing their pharmacokinetic and pharmacodynamic properties but also bioavailability, therapeutic efficacy and toxicity. Therefore, an in-depth understanding of underlying parameters controlling nanocarrier endocytosis and intracellular fate is essential. In order to direct nanoparticles towards specific sub-cellular organelles the physicochemical attributes of nanocarriers can be manipulated. These include particle size, shape and surface charge/chemistry. Restricting the particle size of nanocarriers below 200 nm contributes to internalization via clathrin and caveolae mediated pathways. Similarly, a moderate negative surface potential confers endolysosomal escape and targeting towards mitochondria, endoplasmic reticulum (ER) and Golgi. This review aims to provide an insight into these physicochemical attributes of nanocarriers fabricated using amphiphilic graft copolymers affecting cellular internalization. Fundamental principles understood from experimental studies have been extrapolated to draw a general conclusion for the designing of optimized nanoparticulate drug delivery systems and enhanced intracellular uptake via specific endocytic pathway.

20.
Adv Drug Deliv Rev ; 173: 394-415, 2021 06.
Article in English | MEDLINE | ID: mdl-33831474

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a cluster of several liver diseases like hepatic steatosis, non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver (NAFL), liver fibrosis, and cirrhosis which may eventually progress to liver carcinoma. One of the primary key factors associated with the development and pathogenesis of NAFLD is diabetes mellitus. The present review emphasizes on diabetes-associated development of liver fibrosis and its treatment using different lipid nanoparticles such as stable nucleic acid lipid nanoparticles, liposomes, solid lipid nanoparticles, nanostructured lipid carriers, self-nanoemulsifying drug delivery systems, and conjugates including phospholipid, fatty acid and steroid-based. We have comprehensively described the various pathological and molecular events linking effects of elevated free fatty acid levels, insulin resistance, and diabetes with the pathogenesis of liver fibrosis. Various passive and active targeting strategies explored for targeting hepatic stellate cells, a key target in liver fibrosis, have also been discussed in detail in this review.


Subject(s)
Diabetes Mellitus/drug therapy , Drug Delivery Systems , Hypoglycemic Agents/therapeutic use , Lipids/chemistry , Liver Cirrhosis/drug therapy , Nanoparticles/chemistry , Animals , Diabetes Mellitus/pathology , Drug Carriers/chemistry , Humans , Hypoglycemic Agents/chemistry , Liver Cirrhosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...