Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Hepatology ; 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38950389

ABSTRACT

BACKGROUND AND AIMS: Chronic liver disease leads to ~2 million deaths annually. Cyclic AMP (cAMP) signaling has long been studied in liver injury, particularly in the regulation of fatty acid (FA) ß-oxidation and pro-inflammatory polarization of tissue-resident lymphocytes. Phosphodiesterase 4B inhibition has been explored as a therapeutic modality, but these drugs have had limited success and are known to cause significant adverse effects. The PDE4 inhibitor 2-(4-([2-(5-Chlorothiophen-2-yl)-5-ethyl-6-methylpyrimidin-4-yl]amino)phenyl)acetic acid) (known as A-33) has yet to be explored for the treatment of metabolic diseases. APPROACH AND RESULTS: Herein, we evaluated the efficacy of A-33 in the treatment of animal models of alcohol-associated liver disease and steatotic liver disease. We demonstrated that A-33 effectively ameliorated the signs and symptoms of chronic liver disease, resulting in significant decreases in serum alanine aminotransferase and aspartate aminotransferase levels, decreased overall fat and collagen deposition in the liver, decreased intrahepatic triglyceride concentrations, and normalized expression of genes related to ß-oxidation of fatty acids, inflammation, and extracellular matrix deposition. We also designed and synthesized a novel analog of A-33, termed MDL3, which inhibited both phosphodiesterase 4B and PDE5A and was more effective in ameliorating pathophysiological signs and symptoms of liver injury and inflammation. In addition, MDL3 re-sensitized obese mice to glucose and significantly inhibited the pathological remodeling of adipose tissue, which was not observed with A-33 administration. CONCLUSIONS: In conclusion, we synthesized and demonstrated that MDL3, a novel phosphodiesterase 4B and PDE5A inhibitor, presents a promising avenue of exploration for treating chronic liver disease.

2.
Expert Opin Ther Targets ; : 1-29, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38878273

ABSTRACT

INTRODUCTION: Chronic liver disease (CLD) is a complex disease associated with profound dysfunction. Despite an incredible burden, the first and only pharmacotherapy for metabolic-associated steatohepatitis was only approved in March of this year, indicating a gap in the translation of preclinical studies. There is a body of preclinical work on the application of phosphodiesterase 4 inhibitors in CLD, none of these molecules have been successfully translated into clinical use. AREAS COVERED: To design therapies to combat CLD, it is essential to consider the dysregulation of other tissues that contribute to its development and progression. As such, proper therapies must combat this throughout the body rather than focusing only on the liver. To detail this, literature characterizing the pathogenesis of CLD was pulled from PubMed, with a particular focus placed on the role of PDE4 in inflammation and metabolism. Then, the focus is shifted to detailing the available information on existing PDE4 inhibitors. EXPERT OPINION: This review gives a brief overview of some of the pathologies of organ systems that are distinct from the liver but contribute to disease progression. The demonstrated efficacy of PDE4 inhibitors in other human inflammatory diseases should earn them further examination for the treatment of CLD.

3.
ACS Appl Mater Interfaces ; 16(23): 29686-29698, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38813771

ABSTRACT

Gemcitabine (GEM) is a nucleoside analogue approved as a first line of therapy for pancreatic ductal adenocarcinoma (PDAC). However, rapid metabolism by plasma cytidine deaminase leading to the short half-life, intricate intracellular metabolism, ineffective cell uptake, and swift development of chemoresistance downgrades the clinical efficacy of GEM. ONC201 is a small molecule that inhibits the Akt and ERK pathways and upregulates the TNF-related apoptosis-inducing ligand (TRAIL), which leads to the reversal of both intrinsic and acquired GEM resistance in PDAC treatment. Moreover, the pancreatic cancer cells that were able to bypass apoptosis after treatment of ONC201 get arrested in the G1-phase, which makes them highly sensitive to GEM. To enhance the in vivo stability of GEM, we first synthesized a disulfide bond containing stearate conjugated GEM (lipid-GEM), which makes it sensitive to the redox tumor microenvironment (TME) comprising high glutathione levels. In addition, with the help of colipids 1,2-dioleoyl-glycero-3-phosphocholine (DOPC), cholesterol, and 1,2-distearoyl-glycero-3-phosphoethanolamine-poly(ethylene glycol)-2000 (DSPE-PEG 2000), we were able to synthesize the lipid-GEM conjugate and ONC201 releasing liposomes. A cumulative drug release study confirmed that both ONC201 and GEM showed sustained release from the formulation. Since MUC1 is highly expressed in 70-90% PDAC, we conjugated a MUC1 binding peptide in the liposomes which showed higher cytotoxicity, apoptosis, and cellular internalization by MIA PaCa-2 cells. A biodistribution study further confirmed that the systemic delivery of the liposomes through the tail vein resulted in a higher accumulation of drugs in orthotopic PDAC tumors in NSG mice. The IHC of the excised tumor grafts further confirmed the higher apoptosis and lower metastasis and cell proliferation. Thus, our MUC1 targeting binary drug-releasing liposomal formulation showed higher drug payload, enhanced plasma stability, and accumulation of drugs in the pancreatic orthotopic tumor and thus is a promising therapeutic alternative for the treatment of PDAC.


Subject(s)
Deoxycytidine , Gemcitabine , Pancreatic Neoplasms , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Animals , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Humans , Cell Line, Tumor , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Apoptosis/drug effects , Liposomes/chemistry , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
4.
Cell Death Discov ; 10(1): 158, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553450

ABSTRACT

Approximately 90% of pancreatic cancer (PC) contain KRAS mutations. Mutated KRAS activates the downstream oncogenic PI3K/AKT and MEK signaling pathways and induces drug resistance. However, targeting both pathways with different drugs can also lead to excessive toxicity. ONC201 is a dual PI3K/AKT and MEK pathway inhibitor with an excellent safety profile that targets death receptor 5 (DR5) to induce apoptosis. Gemcitabine (GEM) is a first-line chemotherapy in PC, but it is metabolically unstable and can be stabilized by a prodrug approach. In this study, phospho-Akt, phospho-mTOR, and phospho-ERK protein expressions were evaluated in patient PDAC-tissues (n = 10). We used lipid-gemcitabine (L_GEM) conjugate, which is more stable and enters the cells by passive diffusion. Further, we evaluated the efficacy of L_GEM and ONC201 in PC cells and "KrasLSL-G12D; p53LoxP; Pdx1-CreER (KPC) triple mutant xenograft tumor-bearing mice. PDAC patient tissues showed significantly higher levels of p-AKT (Ser473), p-ERK (T202/T204), and p-mTOR compared to surrounding non-cancerous tissues. ONC201 in combination with L_GEM, showed a superior inhibitory effect on the growth of MIA PaCa-2 cells. In our in-vivo study, we found that ONC201 and L_GEM combination prevented neoplastic proliferation via AKT/ERK blockade to overcome chemoresistance and increased T-cell tumor surveillance. Simultaneous inhibition of the PI3K/AKT and MEK pathways with ONC201 is an attractive approach to potentiate the effect of GEM. Our findings provide insight into rational-directed precision chemo and immunotherapy therapy in PDAC.

6.
J Control Release ; 366: 231-260, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38171473

ABSTRACT

Despite significant treatment efforts, pancreatic ductal adenocarcinoma (PDAC), the deadliest solid tumor, is still incurable in the preclinical stages due to multifacet stroma, dense desmoplasia, and immune regression. Additionally, tumor heterogeneity and metabolic changes are linked to low grade clinical translational outcomes, which has prompted the investigation of the mechanisms underlying chemoresistance and the creation of effective treatment approaches by selectively targeting genetic pathways. Since targeting upstream molecules in first-line oncogenic signaling pathways typically has little clinical impact, downstream signaling pathways have instead been targeted in both preclinical and clinical studies. In this review, we discuss how the complexity of various tumor microenvironment (TME) components and the oncogenic signaling pathways that they are connected to actively contribute to the development and spread of PDAC, as well as the ways that recent therapeutic approaches have been targeted to restore it. We also illustrate how many endogenous stimuli-responsive linker-based nanocarriers have recently been developed for the specific targeting of distinct oncogenes and their downstream signaling cascades as well as their ongoing clinical trials. We also discuss the present challenges, prospects, and difficulties in the development of first-line oncogene-targeting medicines for the treatment of pancreatic cancer patients.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Drug Delivery Systems , Signal Transduction , Tumor Microenvironment
7.
J Control Release ; 369: 684-686, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-37778467
8.
Cancer Lett ; 579: 216462, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37924937

ABSTRACT

Advanced pancreatic cancer patients have a dismal prognosis despite advances in integrative therapy. The field of tumor immunology has witnessed significant advancements for cancer treatment. However, immunotherapy for pancreatic cancer is not very effective due to its highly complex tumor microenvironment (TME). Natural killer (NK) cells are lymphocytes that play an important role in the innate immune system. NK cells do not require antigen pre-sensitization, nor are they confined by the major histocompatibility complex (MHC). NK cells have the potential to eliminate cancer cells through CAR-dependent and CAR-independent pathways, demonstrating reduced levels of systemic toxicity in the process. The availability of several potential sources of NK cells is an additional benefit that contributes to meeting the therapeutic criteria. Adding nanotechnology to enhance the functions of effector NK cells is also an appealing strategy. This article primarily discusses various approaches recently been utilized to enhance the NK functions for the treatment of pancreatic cancer. In addition, new advances in boosting NK cell therapeutic efficacy by nanoparticle mediation are presented, with a focus on pancreatic cancer.


Subject(s)
Nanoparticles , Neoplasms , Pancreatic Neoplasms , Humans , Neoplasms/pathology , Killer Cells, Natural , Immunotherapy , Pancreatic Neoplasms/therapy , Immunotherapy, Adoptive , Tumor Microenvironment
9.
Biomaterials ; 295: 122049, 2023 04.
Article in English | MEDLINE | ID: mdl-36827892

ABSTRACT

Alcohol-associated liver disease (ALD) and its complications are significant health problems worldwide. Several pathways in ALD are influenced by alcohol that drives inflammation, fatty acid metabolism, and fibrosis. Although miR-96 has become a key regulator in several liver diseases, its function in ALD remains unclear. In contrast, sonic hedgehog (SHH) signaling has a well-defined role in liver disease through influencing the activation of hepatic stellate cells (HSCs) and the inducement of liver fibrosis. In this study, we investigated the expression patterns of miR-96 and Hh molecules in mouse and human liver samples. We showed that miR-96 and Shh were upregulated in ethanol-fed mice. Furthermore, alcoholic hepatitis (AH) patient specimens also showed upregulated FOXO3a, TGF-ß1, SHH, and GLI2 proteins. We then examined the effects of Hh inhibitor MDB5 and anti-miR-96 on inflammatory and extracellular matrix (ECM)-related genes. We identified FOXO3 and SMAD7 as direct target genes of miR-96. Inhibition of miR-96 decreased the expression of these genes in vitro in AML12 cells, HSC-T6 cells, and in vivo in ALD mice. Furthermore, MDB5 decreased HSCs activation and the expression of ECM-related genes, such as Gli1, Tgf-ß1, and collagen. Lipid nanoparticles (LNPs) loaded with the combination of MDB5, and anti-miR-96 ameliorated ALD in mice. Our study demonstrated that this combination therapy could serve as a new therapeutic target for ALD.


Subject(s)
MicroRNAs , Transforming Growth Factor beta1 , Animals , Humans , Mice , Antagomirs/pharmacology , Ethanol/adverse effects , Hedgehog Proteins/metabolism , Liver/pathology , Liver Cirrhosis/drug therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Transforming Growth Factor beta1/metabolism
10.
J Control Release ; 354: 80-90, 2023 02.
Article in English | MEDLINE | ID: mdl-36599397

ABSTRACT

Medulloblastoma (MB) is a malignant pediatric brain tumor which shows upregulation of MYC and sonic hedgehog (SHH) signaling. SHH inhibitors face acquired resistance, which is a major cause of relapse. Further, direct MYC oncogene inhibition is challenging, inhibition of MYC upstream insulin-like growth factor/ phosphatidylinositol-4,5-bisphosphate 3-kinase (IGF/PI3K) is a promising alternative. While PI3K inhibition activates resistance mechanisms, simultaneous inhibition of bromodomain-containing protein 4 (BRD4) and PI3K can overcome resistance. We synthesized a new molecule 8-(2,3-dihydrobenzo[b] [1, 4] dioxin-6-yl)-2-morpholino-4H-chromen-4-one (MDP5) that targets both BRD4 and PI3K pathways. We used X-ray crystal structures and a molecular modeling approach to confirm the interactions between MDP5 with bromo domains (BDs) from both BRD2 and BRD4, and molecular modeling for PI3K binding. MDP5 was shown to inhibit target pathways and MB cell growth in vitro and in vivo. MDP5 showed higher potency in DAOY cells (IC50 5.5 µM) compared to SF2523 (IC50 12.6 µM), and its IC50 values in HD-MB03 cells were like SF2523. Treatment of MB cells with MDP5 significantly decreased colony formation, increased apoptosis, and halted cell cycle progression. Further, MDP5 was well tolerated in NSG mice bearing either xenograft or orthotopic MB tumors at the dose of 20 mg/kg, and significantly reduced tumor growth and prolonged animal survival.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Medulloblastoma , Humans , Mice , Animals , Transcription Factors , Nuclear Proteins , Phosphatidylinositol 3-Kinases/metabolism , Hedgehog Proteins , Signal Transduction , Cell Proliferation , Cell Line, Tumor , Cell Cycle Proteins
11.
J Control Release ; 350: 668-687, 2022 10.
Article in English | MEDLINE | ID: mdl-36057395

ABSTRACT

Our body keeps separating the toxic chemicals in the blood from the brain. A significant number of drugs do not enter the central nervous system (CNS) due to the blood-brain barrier (BBB). Certain diseases, such as tumor growth and stroke, are known to increase the permeability of the BBB. However, the heterogeneity of this permeation makes it difficult and unpredictable to transport drugs to the brain. In recent years, research has been directed toward increasing drug penetration inside the brain, and nanomedicine has emerged as a promising approach. Active targeting requires one or more specific ligands on the surface of nanoparticles (NPs), which brain endothelial cells (ECs) recognize, allowing controlled drug delivery compared to conventional targeting strategies. This review highlights the mechanistic insights about different cell types contributing to the development and maintenance of the BBB and summarizes the recent advancement in brain-specific NPs for different pathological conditions. Furthermore, fundamental properties of brain-targeted NPs will be discussed, and the standard lesion features classified by neurological pathology are summarized.


Subject(s)
Endothelial Cells , Nanoparticles , Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Delivery Systems , Endothelial Cells/metabolism , Nanomedicine , Nanoparticles/chemistry , Pharmaceutical Preparations/metabolism
12.
Pharmaceutics ; 14(9)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36145643

ABSTRACT

The incidence of alcoholic liver disease (ALD) is increasing worldwide while no effective treatment has been approved. The progression of ALD has proven to be related to the upregulation of phosphodiesterase 4 (PDE4) expression, and PDE4 inhibitors showed potential to improve ALD. However, the application of PDE4 inhibitors is limited by the gastrointestinal side effects due to PDE4D inhibition. Therefore, we used a novel PDE4B inhibitor KVA-D88 as the therapeutic for ALD treatment. KVA-D88 inhibited inflammatory response, promoted ß-oxidation, increased the level of antioxidants in the hepatocytes, and suppressed hepatic stellate cell (HSC) activation in vitro. To improve the solubility and availability in vivo, KVA-D88 was encapsulated into mPEG-b-P(CB-co-LA) nanoparticles (NPs) by solvent evaporation, with a mean particle size of 135 nm and drug loading of 4.2%. We fed the male C57BL/6 mice with a Lieber-DeCarli liquid diet containing 5% (v/v) ethanol for 6 weeks to induce ALD. Systemic administration of KVA-D88 free drug and KVA-D88-loaded NPs at 5 mg/kg significantly improved the ALD in mice. KVA-D88 significantly ameliorated alcohol-induced hepatic injury and inflammation. KVA-D88 also markedly reduced steatosis by promoting fatty acid ß-oxidation. Liver fibrosis and reactive oxygen species (ROS)-caused cellular damage was observed to be alleviated by KVA-D88. KVA-D88-loaded NPs proved better efficacy than free drug in the animal study. In conclusion, the novel PDE4B inhibitor KVA-D88-loaded NPs have the potential to treat ALD in mice.

13.
J Control Release ; 350: 569-583, 2022 10.
Article in English | MEDLINE | ID: mdl-36037976

ABSTRACT

Microtubule binding agents such as paclitaxel and vincristine have activity in metastatic melanoma. However, even responsive tumors develop resistance, highlighting the need to investigate new drug molecules. Here, we showed that a new compound, CH-2-102, developed by our group, has high anti-tumor efficacy in human and murine melanoma cells. We confirmed that CH-2-102 robustly suppresses the microtubule polymerization process by directly interacting with the colchicine binding site. Our results unveil that CH-2-102 suppresses microtubule polymerization and subsequently induces G2 phase cell arrest as one of the possible mechanisms. Notably, CH-2-102 maintains its efficacy even in the paclitaxel resistance melanoma cells due to different binding sites and a non-Pgp substrate. We developed a pH-responsive drug-polymer Schiff bases linker for high drug loading into nanoparticles (NPs). Our CH-2-102 conjugated NPs induced tumor regression more effectively than Abraxane® (Nab-paclitaxel, N-PTX), free drug, and non-sensitive NPs in B16-F10 cell-derived lung metastasis mouse model. Furthermore, our results suggest that the formulation has a high impact on the in vivo efficacy of the drug and warrants further investigation in other cancers, particularly taxane resistant. In conclusion, the microtubule polymerization inhibitor CH-2-102 conjugated pH-responsive NPs induce tumor regression in lung metastasis melanoma mice, suggesting it may be an effective strategy for treating metastatic melanoma.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Melanoma , Albumin-Bound Paclitaxel/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Colchicine/pharmacology , Colchicine/therapeutic use , Humans , Hydrogen-Ion Concentration , Lung/metabolism , Lung Neoplasms/pathology , Melanoma/pathology , Mice , Nanomedicine , Paclitaxel/chemistry , Polymers/chemistry , Schiff Bases/pharmacology , Schiff Bases/therapeutic use , Tubulin Modulators/chemistry , Tubulin Modulators/therapeutic use , Vincristine
14.
J Geriatr Oncol ; 13(6): 871-874, 2022 07.
Article in English | MEDLINE | ID: mdl-35450817

ABSTRACT

INTRODUCTION: Survival benefit associated with intensive over low-intensity chemotherapy in older adults with acute myeloid leukemia (AML) is controversial. Geriatric assessment and genetic risk categories correlate with survival following intensive chemotherapy in older adults with AML and can guide treatment selection. MATERIALS AND METHODS: In a single-center trial, we integrated both geriatric assessment, and genetic risk categories to personalize selection of intensive versus low-intensity chemotherapy in older adults ≥60 years with AML (NCT03226418). In the present report, we demonstrate feasibility of this approach. RESULTS: Broad eligibility criteria and co-management of patients with community oncologists allowed enrollment of 45% of all patients with AML treated at our center during the study period. The median time from enrollment to therapy initiation was two days (range 0-9). Over half of the trial patients had a score of ≥3 on hematopoietic cell transplantation comorbidity index, impairment in physical function (Short Physical Performance Battery), and Montreal Cognitive Assessment. Three fit patients received intensive chemotherapy, whereas other patients received low-intensity chemotherapy. Mortality at 30 days from diagnosis was 3.7% (95% confidence interval [CI] 0.7-18.3%) and at 90 days was 29.6% (95% CI 15.9-48.5%). One-year overall survival was 66% (95% CI 60-87%). DISCUSSION: Our data demonstrate the feasibility of integrating geriatric assessment in precision oncology trials to define fitness for intensive chemotherapy. Broad eligibility criteria and academic-community collaboration can expand access to clinical trials.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Aged , Geriatric Assessment , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Precision Medicine , Risk Factors , Treatment Outcome
15.
Int J Pharm ; 615: 121470, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35041913

ABSTRACT

In this study, we evaluated the aerodynamic performance, dissolution, and permeation behavior of micronized fluticasone propionate (FP) and magnesium stearate (MgSt) binary mixtures. Micronized FP was dry mixed with 2% w/w MgSt using a tumble mixer and a resonant acoustic mixer (RAM) with and without heating. The mixing efficacy was determined by X-ray powder diffraction (XRPD) and differential scanning calorimetry (DSC) analysis. Additional techniques were used to determine powder properties such as the dynamic vapor sorption (DVS), particle size distribution (PSD) by laser diffraction light scattering, and particle surface properties by scanning electron microscope (SEM). The aerodynamic performance was studied by the next-generation impactor (NGI) using drug-loaded capsules in a PlastiApi® device. Physiochemical properties such as porosity, particle size distribution, and surface area of the formulations were studied with adsorption and desorption curves fitted to several well-known models including Brunauer-Emmett-Teller (BET), Barret Joyner Halenda (BJH), and the density functional theory (DFT). The dissolution behavior of the formulations collected on the transwell inserts incorporated into stages 3, 5, and 7 of the NGI with a membrane providing an air interface was evaluated. Drug permeability of formulations was assessed by directly depositing particles on Calu-3 cells at the air-liquid interface (ALI). Drug concentration was determined by LC-MS/MS. A better MgSt mixing on micronized FP particles was achieved by mixing with a RAM with and without heating than with a tumble mixer. A significant concomitant increase in the % of emitted dose and powder aerosol performance was observed after MgSt mixing. Formulation 4 (RAM mixing at room temperature) showed the highest rate of permeability and correlation with dissolution profile. The results show that the surface enrichment of hydrophobic MgSt improved aerosolization properties and the dissolution and permeability rate of micronized FP by reducing powder agglomerations. A simple low-shear acoustic dry powder mixing method was found to be efficient and substantially improved the powder aerosolization properties and enhanced dissolution and permeability rate.


Subject(s)
Dry Powder Inhalers , Tandem Mass Spectrometry , Administration, Inhalation , Aerosols , Chromatography, Liquid , Fluticasone , Particle Size , Permeability , Powders , Stearic Acids , Surface Properties
16.
Biomaterials ; 278: 121138, 2021 11.
Article in English | MEDLINE | ID: mdl-34634662

ABSTRACT

Chemoresistance and inadequate therapeutics transport across the blood brain barrier (BBB) remain the major barriers to treating medulloblastoma (MB). Hedgehog (Hh) and IGF/PI3K pathways regulate tumor cell proliferation and resistance in MB. Current Hh inhibitors are effective initially to treat SHH-MB but acquire resistance. Herein, we showed that Hh inhibitor MDB5 and BRD4/PI3K dual inhibitor SF2523 synergistically inhibited the proliferation of DAOY and HD-MB03 cells when used in combination. Treatment of these MB cells with the combination of MDB5 and SF2523 significantly decreased colony formation and expression of MYCN, p-AKT, and cyclin D1 but significantly increased in Bax expression, compared to individual drugs. We used our previously reported copolymer mPEG-b-PCC-g-DC copolymer, which showed 8.7 ± 1.0 and 6.5 ± 0.1% loading for MDB5 and SF2523 when formulated into nanoparticles (NPs). There was sustained drug release from NPs, wherein 100% of MDB5 was released in 50 h, but only 60% of SF2523 was released in 80 h. Targeted NPs prepared by mixing 30:70 ratio of COG-133-PEG-b-PBC and mPEG-b-PCC-g-DC copolymer delivered a significantly higher drug concentration in the cerebellum at 6 and 24h after intravenous injection into orthotopic SHH-MB tumor-bearing NSG mice. Moreover, systemic administration of COG-133-NPs loaded with MDB5 and SF2523 resulted in decreased tumor burden compared to non-targeted drug-loaded NPs, without any hepatic toxicity. In conclusion, our nanomedicine of MDB5 and SF2523 offers a novel therapeutic strategy to treat chemoresistant MB.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cerebellar Neoplasms , Medulloblastoma , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzene Derivatives , Cell Line, Tumor , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Drug Synergism , Hedgehog Proteins , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Mice , Morpholines , Nanomedicine , Nuclear Proteins , Phosphatidylinositol 3-Kinases , Pyrans , Pyridines , Transcription Factors
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.
Adv Drug Deliv Rev ; 176: 113888, 2021 09.
Article in English | MEDLINE | ID: mdl-34314787

ABSTRACT

Type 2 diabetes mellitus (T2DM) associated non-alcoholic fatty liver disease (NAFLD) is the fourth-leading cause of death. Hyperglycemia induces various complications, including nephropathy, cirrhosis and eventually hepatocellular carcinoma (HCC). There are several etiological factors leading to liver disease development, which involve insulin resistance and oxidative stress. Free fatty acid (FFA) accumulation in the liver exerts oxidative and endoplasmic reticulum (ER) stresses. Hepatocyte injury induces release of inflammatory cytokines from Kupffer cells (KCs), which are responsible for activating hepatic stellate cells (HSCs). In this review, we will discuss various molecular targets for treating chronic liver diseases, including homeostasis of FFA, lipid metabolism, and decrease in hepatocyte apoptosis, role of growth factors, and regulation of epithelial-to-mesenchymal transition (EMT) and HSC activation. This review will also critically assess different strategies to enhance drug delivery to different cell types. Targeting nanocarriers to specific liver cell types have the potential to increase efficacy and suppress off-target effects.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Delivery Systems , Liver Cirrhosis/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Humans , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism
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