Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-39009933

ABSTRACT

Glioblastoma (GBM) is the deadliest adult brain cancer. The current standard-of-care chemotherapy using orally administered temozolomide (TMZ) presents poor improvement in patient survival, emphasizing the compelling need for new therapies. A possible chemotherapeutic alternative is docetaxel (DTX), which possesses higher tumoricidal potency against GBM cells. However, its limited blood-brain barrier (BBB) permeability poses a constraint on its application. Nonetheless, nanomedicine offers promising avenues for overcoming this challenge. Angiopep-2 (ANG2) is a peptide that targets the BBB-overexpressed low-density lipoprotein receptor (LDLR). In this work, we managed, for the first time, to employ a pioneering approach of covalently linking zein protein with polyethylene glycol (PEG) and ANG2 prior to its formulation into nanoparticles (ZNPs) with enhanced stability and LDLR-mediated brain targetability, respectively. Carbodiimide and click chemistry approaches were optimized, resulting in functional modification of zein with around 25% PEG, followed by functional modification of PEG with nearly 100% ANG2. DTX-loaded ZNPs presented 100 nm average size, indicating high suitability for BBB crossing through receptor-mediated transcytosis. ZNPs maintained the cytotoxic effect of the loaded DTX against GBM cells, while demonstrating a safe matrix against BBB cells. Importantly, these brain-targeted ZNPs showcased up to fourfold enhancement in blood-to-brain permeability in a BBB in vitro model, highlighting the potential of this novel approach of BBB targeting in significantly improving therapeutic outcomes for GBM patients. The versatility of the system and the possibility of significantly increasing drug concentration in the brain open the door to its future application in a wide range of other brain-related diseases.

2.
Int J Pharm ; 655: 124004, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38492899

ABSTRACT

Photodynamic therapy (PDT) is a suitable alternative to currently employed cancer treatments. However, the hydrophobicity of most photosensitizers (e.g., zinc phthalocyanine (ZnPC)) leads to their aggregation in blood. Moreover, non-specific accumulation in skin and low clearance rate of ZnPC leads to long-lasting skin photosensitization, forcing patients with a short life expectancy to remain indoors. Consequently, the clinical implementation of these photosensitizers is limited. Here, benzyl-poly(ε-caprolactone)-b-poly(ethylene glycol) micelles encapsulating ZnPC (ZnPC-M) were investigated to increase the solubility of ZnPC and its specificity towards cancers cells. Asymmetric flow field-flow fractionation was used to characterize micelles with different ZnPC-to-polymer ratios and their stability in human plasma. The ZnPC-M with the lowest payload (0.2 and 0.4% ZnPC w/w) were the most stable in plasma, exhibiting minimal ZnPC transfer to lipoproteins, and induced the highest phototoxicity in three cancer cell lines. Nanobodies (Nbs) with binding specificity towards hepatocyte growth factor receptor (MET) or epidermal growth factor receptor (EGFR) were conjugated to ZnPC-M to facilitate cell targeting and internalization. MET- and EGFR-targeting micelles enhanced the association and the phototoxicity in cells expressing the target receptor. Altogether, these results indicate that ZnPC-M decorated with Nbs targeting overexpressed proteins on cancer cells may provide a better alternative to currently approved formulations.


Subject(s)
Isoindoles , Organometallic Compounds , Photochemotherapy , Humans , Photosensitizing Agents/chemistry , Micelles , Polymers , Photochemotherapy/methods , Zinc Compounds , Organometallic Compounds/pharmacology , Organometallic Compounds/chemistry , ErbB Receptors , Cell Line, Tumor
3.
Nanomedicine (Lond) ; 19(7): 633-651, 2024 03.
Article in English | MEDLINE | ID: mdl-38445583

ABSTRACT

Nanomedicine has opened up new avenues for cancer treatment by enhancing drug solubility, permeability and targeted delivery to cancer cells. Despite its numerous advantages over conventional therapies, nanomedicine may exhibit off-target drug distribution, harming nontarget regions. The increased permeation and retention effect of nanomedicine in tumor sites also has its limitations, as abnormal tumor vasculature, dense stroma structure and altered tumor microenvironment (TME) may result in limited intratumor distribution and therapeutic failure. However, TME-responsive nanomedicine has exhibited immense potential for efficient, safe and precise delivery of therapeutics utilizing stimuli specific to the TME. This review discusses the mechanistic aspects of various TME-responsive biopolymers and their application in developing various types of TME-responsive nanomedicine.


Subject(s)
Nanomedicine , Neoplasms , Humans , Tumor Microenvironment , Neoplasms/drug therapy , Neoplasms/pathology , Drug Delivery Systems
4.
Adv Drug Deliv Rev ; 207: 115204, 2024 04.
Article in English | MEDLINE | ID: mdl-38342241

ABSTRACT

Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional "polarization" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.


Subject(s)
Anti-Inflammatory Agents , Macrophages , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/metabolism , Inflammation/metabolism , Wound Healing , Signal Transduction
5.
Macromol Biosci ; 23(9): e2300077, 2023 09.
Article in English | MEDLINE | ID: mdl-37163974

ABSTRACT

Evodiamine (Evo) is a natural, biologically active plant alkaloid with wide range of pharmacological activities. In the present study Evo-loaded folate-conjugated Pluronic F108 nano-micelles (ENM) is synthesized to enhance the therapeutic efficacy of Evo against cervical cancer. ENM are synthesized, physicochemically characterized and in vitro anticancer activity is performed. The study demonstrates that ENM have nanoscale size (50.33 ± 3.09 nm), monodispersity of 0.122 ± 0.072, with high drug encapsulation efficiency (71.30 ± 3.76%) and controlled drug release at the tumor microenvironment. ENM showed dose-dependent and time-dependent cytotoxicity against HeLa human cervical cancer cells. The results of in vitro anticancer studies demonstrated that ENM have significant anticancer effects and greatly induce apoptosis as compared to pure Evo. The cellular uptake study suggests that increased anticancer activity of ENM is due to the improved intracellular delivery of Evo through overexpressed folate receptors. Overall, the designed ENM can be a potential targeted delivery system for hydrophobic anticancer bioactive compound like Evo.


Subject(s)
Poloxamer , Uterine Cervical Neoplasms , Female , Humans , Poloxamer/chemistry , Uterine Cervical Neoplasms/drug therapy , Folic Acid/chemistry , Micelles , Apoptosis , Cell Death , Cell Line, Tumor , Drug Carriers/pharmacology , Drug Carriers/chemistry , Tumor Microenvironment
6.
Mol Pharm ; 20(1): 663-679, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36413707

ABSTRACT

Rutin (RUT) is a flavonoid obtained from a natural source and is reported for antithrombotic potential, but its delivery remains challenging because of its poor solubility and bioavailability. In this research, we have fabricated novel rutin loaded liposomes (RUT-LIPO, nontargeted), liposomes conjugated with RGD peptide (RGD-RUT-LIPO, targeted), and abciximab (ABX-RUT-LIPO, targeted) by ethanol injection method. The particle size, ζ potential, and morphology of prepared liposomes were analyzed by using DLS, SEM, and TEM techniques. The conjugation of targeting moiety on the surface of targeted liposomes was confirmed by XPS analysis and Bradford assay. In vitro assessment such as blood clot assay, aPTT assay, PT assay, and platelet aggregation analysis was performed using human blood which showed the superior antithrombotic potential of ABX-RUT-LIPO and RGD-RUT-LIPO liposomes. The clot targeting efficiency was evaluated by in vitro imaging and confocal laser scanning microscopy. A significant (P < 0.05) rise in the affinity of targeted liposomes toward activated platelets was demonstrated that revealed their remarkable potential in inhibiting thrombus formation. Furthermore, an in vivo study executed on Sprague Dawley rats (FeCl3 model) demonstrated improved antithrombotic activity of RGD-RUT-LIPO and ABX-RUT-LIPO compared with pure drug. The pharmacokinetic study performed on rats demonstrates the increase in bioavailability when administered as liposomal formulation as compared to RUT. Moreover, the tail bleeding assay and clotting time study (Swiss Albino mice) indicated a better antithrombotic efficacy of targeted liposomes than control preparations. Additionally, biocompatibility of liposomal formulations was determined by an in vitro hemolysis study and cytotoxicity assay, which showed that they were hemocompatible and safe for human use. A histopathology study on rats suggested no severe toxicity of prepared liposomal formulations. Thus, RUT encapsulated nontargeted and targeted liposomes exhibited superior antithrombotic potential over RUT and could be used as a promising carrier for future use.


Subject(s)
Liposomes , Thrombosis , Mice , Rats , Humans , Animals , Drug Delivery Systems/methods , Fibrinolytic Agents/pharmacology , Rutin , Rats, Sprague-Dawley , Oligopeptides , Thrombosis/drug therapy
7.
Int J Pharm X ; 4: 100139, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36420371

ABSTRACT

Apoptosis is an important process that directly affects the response of cancer cells to anticancer drugs. Among different factors involved in this process, the BcL-xL protein plays a critical role in inhibiting apoptosis induced by chemotherapy agents. Henceforth, its downregulation may have a synergistic activity that lowers the necessary dose of anticancer agents. In this study, anti-Bcl-xL siRNA were formulated within an EGFR-targeted nanomedicine with scFv ligands (NM-scFv) and its activity was tested in the non-small cell lung cancer (NSCLC) cell line H460. The obtained NMs-scFv anti-Bcl-xL were suitable for intravenous injection with sizes around 100 nm, a high monodispersity level and good siRNA complexation capacity. The nanocomplex's functionalization with anti-EGFR scFv ligands was shown to allow an active gene delivery into H460 cells and led to approximately 63% of gene silencing at both mRNA and protein levels. The NM-scFv anti-Bcl-xL improved the apoptotic activity of cisplatin and reduced the cisplatin IC50 value in H460 cells by a factor of around three from 0.68 ± 0.12 µM to 2.21 ± 0.18 µM (p < 0.01), respectively, in comparison to that of NM-scFv formulated with control siRNA (p > 0.05).

8.
Nanomedicine (Lond) ; 17(23): 1779-1798, 2022 10.
Article in English | MEDLINE | ID: mdl-36636930

ABSTRACT

Lung cancer is the second leading cause of cancer-related mortality globally, and non-small-cell lung cancer accounts for most lung cancer cases. Nanotechnology-based drug-delivery systems have exhibited immense potential in lung cancer therapy due to their fascinating physicochemical characteristics, in vivo stability, bioavailability, prolonged and targeted delivery, gastrointestinal absorption and therapeutic efficiency of their numerous chemotherapeutic agents. However, traditional chemotherapeutics have systemic toxicity issues; therefore, dietary polyphenols might potentially replace them in lung cancer treatment. Polyphenol-based targeted nanotherapeutics have demonstrated interaction with a multitude of protein targets and cellular signaling pathways that affect major cellular processes. This review summarizes the various molecular mechanisms and targeted therapeutic potentials of nanoengineered dietary polyphenols in the effective management of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma , Lung Neoplasms , Humans , Lung Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Polyphenols/therapeutic use , Polyphenols/chemistry , Lung , Carcinoma/drug therapy
9.
Int J Pharm ; 602: 120652, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33915187

ABSTRACT

Novel glutathione (GSH) redox-sensitive thiolated vitaminE-PEG1000-succinate (TPGH-SH) was synthesized by conjugating TPGS with 4-amino thiophenol (4-ATP) and confirmed by FTIR and NMR studies. Following, docetaxel (DTX) loaded, cetuximab (CTB) conjugated redox sensitive TPGS-SH nanoparticles (TPGS-SH NP) were prepared by dialysis method and screened for size, charge, DTX entrapment, which revealed that size, surface charge and percent entrapment are in the range of 183-227 nm, +18 to +26 mV and 68-71%. SEM, TEM, AFM have reflected the spherical and uniform size of NP with a smooth surface. In-vitro release studies were performed in media containing different concentrations of GSH to study their effect on drug release and drug release of up to 94.5%, at pH 5.5, GSH 20 mM, is observed within 24 h. The pH/redox sensitivity studies revealed the better stability of NP at higher pH and lower GSH concentrations. In-vitro cytotoxicity, cellular uptake, migration and apoptotic assays, performed on A549 cells, have proved that targeted formulation produced higher cytotoxicity (significantly less IC50 value) and uptake and also prevented cell migration. Pharmacokinetic and histopathological screening were performed on CF rats, which demonstrated promising results. The in-vivo efficacy studies on benzo(a)pyrene induced mice lung cancer model showed that targeted TPGS-SH NP has significantly reduced the cell number than the model control.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Nanoparticles , Animals , Cell Line, Tumor , ErbB Receptors , Lung Neoplasms/drug therapy , Mice , Oxidation-Reduction , Particle Size , Polyethylene Glycols , Rats , Vitamin E
10.
J Control Release ; 332: 109-126, 2021 04 10.
Article in English | MEDLINE | ID: mdl-33571549

ABSTRACT

Cancer immunotherapy has revolutionized the standard of care for solid tumors in multiple disease sites. In light of this, immune checkpoint blockade, directly interfering with various immunosuppressive mechanisms in tumor sites, has been actively studied. Inhibitors of cytotoxic T-lymphocyte-associated protein 4 (CTLA 4) and programmed cell death 1 receptor (PD-1) / programmed cell death receptor ligand 1 (PD-L1) could successfully increase survival rate in patients with advanced cancers including melanoma and non-small cell lung cancer, leading to increased survival and different patterns of response including durable response and pseudo-progression. Despite continued development of the immune checkpoint blockades, however, fairly low overall response rate (ORR) levels have been reported for patients with various types of cancer. Fewer than 13% of patients with cancer were reported to respond to immune checkpoint blockades and some patients were diagnosed with severe immune-related adverse events (irAEs). The tumor microenvironment, controlled via various components and mediators, is regarded as the primary cause responsible for failure of immune checkpoint blockades in clinical investigations. In fact, there has been a clinical report that epidermal growth factor receptor (EGFR) inhibition in tumor microenvironment enhanced ORR of PD-1 inhibitors for 29.7% in EGFR-mutated non-small-cell lung carcinoma (NSCLC) patients. Therefore, to enhance the effectiveness and reduce adverse effects of immune checkpoint blockades, the majority of studies have focused on targeting and suppressing the immunosuppressive characteristics of the tumor microenvironment. Herein, we review the components and mediators of tumor microenvironment responsible for failure of immune checkpoint blockades and introduce the recent approaches of tumor microenvironment component-targeted nanomedicine delivery capable of enhancing the efficacy of immune checkpoint blockades. Understanding the active targeting candidates of tumor microenvironment components and the associated treatment strategies could offer insights into the development of combination therapeutics boosting immune checkpoint blockades for clinical applications.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Immune Checkpoint Inhibitors , Immunotherapy , Lung Neoplasms/drug therapy , Nanomedicine , Tumor Microenvironment
11.
Mater Sci Eng C Mater Biol Appl ; 120: 111652, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33545820

ABSTRACT

This work was aimed to formulate transferrin (Tf) receptor targeted gold based theranostic liposomes which contain both docetaxel (DCX) and glutathione reduced gold nanoparticles (AuGSH) for brain-targeted drug delivery and imaging. AuGSH was prepared by reducing chloroauric acid salt using glutathione. The co-loading of DCX and AuGSH into liposomes was achieved by the solvent injection technique, and Tf was post-conjugated on the surface of the liposomes using carboxylated Vit-E TPGS (TPGS-COOH) as a linker. The liposomes were characterized for various parameters such as size, shape, surface charge, and drug release. The Tf receptor targeted gold liposomes were evaluated for the cytotoxicity by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) based colorimetric assay and in-vitro qualitative cellular uptake studies using confocal microscopy. The in-vivo site specific delivery of DCX was analyzed by the brain distribution study of DCX in comparison with marketed formulation (Docel™). A sustained drug release of about 70% was observed from liposomes in the span of 72 h. The in-vivo results demonstrated that targeted gold liposomes were able to deliver DCX into the brain by 3.70, 2.74 and 4.08-folds higher than Docel™ after 30, 120 and 240 min of the treatment, respectively. Besides, the results of these studies have suggested the feasibility of Tf decorated AuGSH and DCX co-loaded liposomes as a promising platform for targeted nano-theranostics.


Subject(s)
Liposomes , Metal Nanoparticles , Brain , Cell Line, Tumor , Drug Carriers , Drug Delivery Systems , Gold , Kinetics
12.
ACS Nano ; 15(1): 1331-1341, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33395272

ABSTRACT

The biofunctionalization of particles with specific targeting moieties forms the foundation for molecular recognition in biomedical applications such as targeted nanomedicine and particle-based biosensing. To achieve a high precision of targeting for nanomedicine and high precision of sensing for biosensing, it is important to understand the consequences of heterogeneities of particle properties. Here, we present a comprehensive methodology to study with experiments and simulations the collective consequences of particle heterogeneities on multiple length scales, called superpositional heterogeneities, in generating reactivity variability per particle. Single-molecule techniques are used to quantify stochastic, interparticle, and intraparticle variabilities, in order to show how these variabilities collectively contribute to reactivity variability per particle, and how the influence of each contributor changes as a function of the system parameters such as particle interaction area, the particle size, the targeting moiety density, and the number of particles. The results give insights into the consequences of superpositional heterogeneities for the reactivity variability in biomedical applications and give guidelines on how the precision can be optimized in the presence of multiple independent sources of variability.

13.
Adv Sci (Weinh) ; 7(13): 2000487, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32670766

ABSTRACT

Acute myelogenous leukemia (AML) is a fatal blood cancer with high patient mortality. Daunorubicin and cytarabine are first-line chemotherapy for AML, with bone marrow transplantation in most cases. Recently, cancer immunotherapy has been challenged in AML and leukemia-niche myeloid cells are promising targets for the AML immunotherapy. Heme oxygenase 1 (HO1) is an antioxidative and cytoprotective enzyme inducing chemo-resistant AML and has been focused as an immune checkpoint molecule in tumor microenvironments. Herein, lipid-polymer hybrid nanoparticle (hNP) is loaded with tin mesoporphyrin (SnMP), a HO1-inhibitor, and non-covalently modified with an engineered antibody for leukemic cell-targeted delivery. HO1-inhibiting T-hNP (T-hNP/SnMP) enhances chemo-sensitivity in human leukemia cells. In a human AML-bearing orthotopic mouse model, intravenously injected T-hNP not only actively targets to human leukemia cells but passively targets to CD11b+ myeloid cells in a bone marrow niche. The T-hNP/SnMP enhances the chemo-therapeutic effect of daunorubicin and boosts immune response by reprogramming bone marrow myeloid cells resulting from the recruitment of the monocyte-lineage and induction of inflammatory genes. The ex vivo study demonstrates an enhanced immune response of HO1-inhibited bone marrow CD11b+ myeloid cells against apoptotic leukemia cells. Collectively, HO1-inhibiting dual cell-targeted T-hNP/SnMP has a strong potential as a novel therapeutic in AML.

14.
Nano Lett ; 19(3): 2128-2137, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30799620

ABSTRACT

Gallium and gallium-based alloys, typical types of liquid metals with unique physiochemical properties, are emerging as a next generation of functional materials in versatile biomedical applications. However, the exploration of their biomedical performance is currently insufficient, and their intrinsic low oxidative resistance is a key factor blocking their further clinical translation. Herein, we report on the surface engineering of liquid metal-based nanoplatforms by an inorganic silica nanoshell based on a novel but facile sonochemical synthesis for highly efficient, targeted, and near-infrared (NIR)-triggered photothermal tumor hyperthermia in the NIR-II biowindow. The inorganic silica-shell engineering of liquid metal significantly enhances the photothermal performance of the liquid metal core as reflected by enhanced NIR absorption, improved photothermal stability by oxidation protection, and abundant surface chemistry for surface-targeted engineering to achieve enhanced tumor accumulation. Systematic in vitro cell-level evaluation and in vivo tumor xenograft assessment demonstrate that (Arg-Gly-Asp) RGD-targeted and silica-coated nanoscale liquid metal substantially induces phototriggered cancer-cell death and photothermal tumor eradication, accompanied by high in vivo biocompatibility and easy excretion out of the body. This work provides the first paradigm for surface-inorganic engineering of liquid metal-based nanoplatforms for achieving multiple desirable therapeutic performances, especially for combating cancer.


Subject(s)
Hyperthermia, Induced/methods , Nanoshells/chemistry , Neoplasms/therapy , Oxidative Stress/drug effects , Alloys/chemical synthesis , Alloys/chemistry , Alloys/pharmacology , Gallium/chemistry , Gallium/pharmacology , Humans , Inorganic Chemicals/chemistry , Ionic Liquids/chemistry , Ionic Liquids/therapeutic use , Nanoshells/therapeutic use , Silicon Dioxide/chemistry
15.
Colloids Surf B Biointerfaces ; 173: 366-377, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30316083

ABSTRACT

Breast cancer, up-regulated with human epidermal growth factor receptor type-2 (HER-2) has led to the concept of developing HER-2 targeted anticancer therapeutics. Docetaxel-loaded D-α-tocopherol polyethylene glycol 1000 succinate conjugated chitosan (TPGS-g-chitosan) nanoparticles were prepared with or without Trastuzumab decoration. The particle size and entrapment efficiency of conventional, non-targeted as well as targeted nanoparticles were in the range of 126-186 nm and 74-78% respectively. In-vitro studies on SK-BR-3 cells showed that docetaxel-loaded non-targeted and HER-2 receptor targeted TPGS-g-chitosan nanoparticles have enhanced the cellular uptake and cytotoxicity with a promising bioadhesion property, in comparison to conventional formulation i.e., Docel™. The IC50 values of non-targeted and targeted nanoparticles from cytotoxic assay were found to be 43 and 223 folds higher than Docel™. The in-vivo pharmacokinetic study showed 2.33, and 2.82-fold enhancement in relative bioavailability of docetaxel for non-targeted and HER-2 receptor targeted nanoparticles, respectively than Docel™. Further, after i.v administration, non-targeted and targeted nanoparticles achieved 3.48 and 5.94 times prolonged half-life in comparison to Docel™. The area under the curve (AUC), relative bioavailability (FR) and mean residence time (MRT) were found to be higher for non-targeted and targeted nanoparticles when compared to Docel™. The histopathology studies of non-targeted and targeted nanoparticles showed less toxicity on vital organs such as lungs, liver, and kidney when compared to Docel™.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Docetaxel/pharmacology , Glycoconjugates/pharmacology , Trastuzumab/pharmacology , Vitamin E/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Availability , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chitosan/chemistry , Docetaxel/chemistry , Docetaxel/pharmacokinetics , Drug Carriers , Female , Glycoconjugates/chemistry , Glycoconjugates/pharmacokinetics , Humans , Kidney/drug effects , Liver/drug effects , Lung/drug effects , Molecular Targeted Therapy , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Rats , Receptor, ErbB-2/metabolism , Trastuzumab/chemistry , Trastuzumab/pharmacokinetics , Tumor Burden/drug effects , Vitamin E/chemistry , Vitamin E/pharmacokinetics , Xenograft Model Antitumor Assays
16.
J Chem Neuroanat ; 95: 29-42, 2019 01.
Article in English | MEDLINE | ID: mdl-29269015

ABSTRACT

Mitochondria, the powerhouse of the neural cells in the brain, are also the seat of certain essential gene signaling pathways that control neuronal functions. Deterioration of mitochondrial functions has been widely reported in normal aging as well as in a spectrum of age-associated neurological diseases, including Parkinson's disease (PD). Evidences accumulated in the recent past provide not only advanced information on the causes of mitochondrial bioenergetics defects and redox imbalance in PD brains, but also much insight into mitochondrial biogenesis, quality control of mitochondrial proteins, and genes, which regulate intra- and extra-mitochondrial signaling that control the general health of neural cells. The mitochondrial quality control machinery is affected in aging and especially in PD, thus affecting intraneuronal protein transport and degradation, which are primarily responsible for accumulation of misfolded proteins and mitochondrial damage in sporadic as well as familial PD. Essentially we considered in the first half of this review, mitochondria-based targets such as mitochondrial oxidative stress and mitochondrial quality control pathways in PD, relevance of mitochondrial DNA mutations, mitophagy, mitochondrial proteases, mitochondrial flux, and finally mitochondria-based therapies possible for PD. Therapeutic aspects are considered in the later half and mitochondria-targeted antioxidant therapy, mitophagy enhancers, mitochondrial biogenesis boasters, mitochondrial dynamics modulators, and gene-based therapeutic approaches are discussed. The present review is a critical assessment of this information to distinguish some exemplary mitochondrial therapeutic targets, and provides a utilitarian perception of some avenues for therapeutic designs on identified mitochondrial targets for PD, a very incapacitating disorder of the geriatric population, world over.


Subject(s)
Aging/physiology , Brain/physiology , Mitochondria/physiology , Parkinson Disease/physiopathology , Aging/pathology , Animals , Brain/pathology , Humans , Mitochondria/pathology
17.
ACS Appl Bio Mater ; 2(8): 3312-3320, 2019 Aug 19.
Article in English | MEDLINE | ID: mdl-35030773

ABSTRACT

Selective tissue visualization and localized tumor regression without affecting the surrounding healthy tissues are critical concerns in cancer nanomedicine. Importantly, the complete wrapping of a flimsy matrix like liposome by multifunctional graphene oxide is an interesting engineering idea for nanomedicine design. Moreover, designing a safe and biodegradable nanohybrid with significant theranostic ability is a current need for targeted combined therapies. Here, we report a comprehensive result of in vivo tumor diagnosis and phototriggered tumor regression using a biodegradable red emissive nanotheranostic system, viz., graphene oxide flakes fortified liposome (GOF-Lipo), functionalized with folic acid (FA): GOF-Lipo-FA. Graphene oxide support enhances the stability of drug-loaded liposomes in an extracellular environment that prevents the premature release of loaded anticancer drug from the liposomal cavity. Promising outcomes of tumor regression (∼300 to 25 mm3) from organized cellular and animal studies are demonstrated in this work. These studies reveal superior biocompatibility, deep intracellular localization, 4T1 breast tumor diagnosis, and long time tumor binding ability of an injected emissive nanohybrid. Overall, a single dose of designed multifunctional systems demonstrates the best tumor regression.

18.
Int J Hyperthermia ; 34(1): 19-29, 2018 02.
Article in English | MEDLINE | ID: mdl-28540812

ABSTRACT

PURPOSE: To demonstrate delivery of Au nanocages to cells using the galectin-1 binding peptide anginex (Ax) and to demonstrate the value of this targeting for selective in vitro photothermal cell killing. MATERIALS AND METHODS: Au nanocages were synthesised, coated with polydopamine (PDA), and conjugated with Ax. Tumour and endothelial cell viability was measured with and without laser irradiation. Photoacoustic (PA) mapping and PA flow cytometry were used to confirm cell targeting in vitro and in tissue slices ex vivo. RESULTS: Cell viability was maintained at ≥50% at 100 pM suggesting low toxicity of the nanocage alone. Combining the targeted construct (25 pM) with low power 808 nm laser irradiation for 10-20 min (a duration previously shown to induce rapid and sustained heating of Au nanocages [AuNC] in solution), resulted in over 50% killing of endothelial and tumour cells. In contrast, the untargeted construct combined with laser irradiation resulted in negligible cell killing. We estimate approximately 6 × 104 peptides were conjugated to each nanocage, which also resulted in inhibition of cell migration. Binding of the targeted nanocage reached a plateau after three hours, and cell association was 20-fold higher than non-targeted nanocages both in vitro and ex vivo on tumour tissue slices. A threefold increase in tumour accumulation was observed in preliminary in vivo studies. CONCLUSIONS: These studies demonstrate Ax's potential as an effective targeting agent for Au-based theranostics to tumour and endothelial cells, enabling photothermal killing. This platform further suggests potential for multimodal in vivo therapy via next-generation drug-loaded nanocages.


Subject(s)
Galectin 1/metabolism , Gold/metabolism , Nanostructures/chemistry , Phototherapy/methods , Animals , Mice , Mice, Inbred BALB C
19.
J Control Release ; 243: 333-341, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27984104

ABSTRACT

An important limitation to successful cancer treatment with chemotherapeutics is the inability to achieve therapeutically effective drug concentrations while avoiding healthy tissue damage. In this work, a new tumor-targeting peptide iRGD (CCRGDKGPDC) was used to modify drug-loaded low temperature-sensitive liposomes (iRGD-LTSL-DOX) to explore the anti-tumor effects in combination with high intensity focused ultrasound (HIFU) in vitro and in vivo. iRGD-LTSL-DOX can specifically target to ανß3-positive cells and locally release the encapsulated doxorubicin (DOX) in a hyperthermia-triggered manner. In vivo results showed that DOX from iRGD-LTSL-DOX was intravascularly released and rapidly penetrated into tumor interstitial space after HIFU-triggered heat treatment, thereby overcoming the limited tumor penetration of anticancer drugs. Significantly stronger anti-tumor efficacy further supported the effective combination of iRGD-LTSL-DOX with HIFU-induced hyperthermia. Our study provided a novel tumor-targeting LTSL-DOX and demonstrated its usefulness in HIFU-induced hyperthermia-triggered drug delivery.


Subject(s)
Doxorubicin/administration & dosage , Drug Delivery Systems , High-Intensity Focused Ultrasound Ablation/methods , Oligopeptides/chemistry , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Hot Temperature , Humans , Liposomes , Mice , Mice, Inbred BALB C , Nanoparticles , Neoplasms/drug therapy , Neoplasms/pathology , Tissue Distribution
20.
Colloids Surf B Biointerfaces ; 147: 129-141, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27497076

ABSTRACT

The aim of this work was to formulate RGD-TPGS decorated theranostic liposomes, which contain both docetaxel (DTX) and quantum dots (QDs) for brain cancer imaging and therapy. RGD conjugated TPGS (RGD-TPGS) was synthesized and conjugation was confirmed by Fourier transform infrared (FTIR) spectroscopy and electrospray ionisation (ESI) mass spectroscopy (ESI-MS). The theranostic liposomes were prepared by the solvent injection method and characterized for their particle size, polydispersity, zeta-potential, surface morphology, drug encapsulation efficiency, and in-vitro release study. Biocompatibility and safety of theranostic liposomes were studied by reactive oxygen species (ROS) generation study and histopathology of brain. In-vivo study was performed for determination of brain theranostic effects in comparison with marketed formulation (Docel™) and free QDs. The particle sizes of the non-targeted and targeted theranostic liposomes were found in between 100 and 200nm. About 70% of drug encapsulation efficiency was achieved with liposomes. The drug release from RGD-TPGS decorated liposomes was sustained for more than 72h with 80% of drug release. The in-vivo results demonstrated that RGD-TPGS decorated theranostic liposomes were 6.47- and 6.98-fold more effective than Docel™ after 2h and 4h treatments, respectively. Further, RGD-TPGS decorated theranostic liposomes has reduced ROS generation effectively, and did not show any signs of brain damage or edema in brain histopathology. The results of this study have indicated that RGD-TPGS decorated theranostic liposomes are promising carrier for brain theranostics.


Subject(s)
Brain/drug effects , Drug Carriers , Drug Delivery Systems , Liposomes/chemistry , Taxoids/pharmacology , Theranostic Nanomedicine , Vitamin E/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Docetaxel , Drug Liberation , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Quantum Dots , Rats , Rats, Inbred Strains , Reactive Oxygen Species/metabolism , Taxoids/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL