Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cancer Sci ; 115(4): 1129-1140, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38351514

RESUMO

Oncolytic viruses (OVs) possess the unique ability to selectively replicate within tumor cells, leading to their destruction, while also reversing the immunosuppression within the tumor microenvironment and triggering an antitumor immune response. As a result, OVs have emerged as one of the most promising approaches in cancer therapy. However, the effective delivery of intravenously administered OVs faces significant challenges imposed by various immune cells within the peripheral blood, hindering their access to tumor sites. Notably, neutrophils, the predominant white blood cell population comprising approximately 50%-70% of circulating white cells in humans, show phagocytic properties. Our investigation revealed that the majority of oncolytic vaccinia viruses (VV) are engulfed and degraded by neutrophils in the bloodstream. The depletion of neutrophils using the anti-LY6G Ab (1-A8) resulted in an increased accumulation of circulating oncolytic VV in the peripheral blood and enhanced deposition at the tumor site, consequently amplifying the antitumor effect. Neutrophils heavily rely on PI3K signaling to sustain their phagocytic process. Additionally, our study determined that the inhibition of the PI3Kinase delta isoform by idelalisib (CAL-101) suppressed the uptake of oncolytic VV by neutrophils. This inhibition led to a greater presence of oncolytic VV in both the peripheral blood and at the tumor site, resulting in improved efficacy against the tumor. In conclusion, our study showed that inhibiting neutrophil functions can significantly enhance the antitumor efficacy of intravenous oncolytic VV.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Vírus Oncolíticos/fisiologia , Vaccinia virus/fisiologia , Neutrófilos/patologia , Terapia Viral Oncolítica/métodos , Fosfatidilinositol 3-Quinases , Neoplasias/patologia , Microambiente Tumoral
2.
Int J Mol Sci ; 24(3)2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-36768907

RESUMO

The variable success in the outcome of randomised controlled trials supplementing coenzyme Q10 (CoQ10) may in turn be associated with a number of currently unresolved issues relating to CoQ10 metabolism. In this article, we have reviewed what is currently known about these factors and where gaps in knowledge exist that need to be further elucidated. Issues addressed include (i) whether the bioavailability of CoQ10 could be improved; (ii) whether CoQ10 could be administered intravenously; (iii) whether CoQ10 could be administered via alternative routes; (iv) whether CoQ10 can cross the blood-brain barrier; (v) how CoQ10 is transported into and within target cells; (vi) why some clinical trials supplementing CoQ10 may have been unsuccessful; and (vii) which is the most appropriate tissue for the clinical assessment of CoQ10 status.


Assuntos
Antioxidantes , Ubiquinona , Ubiquinona/metabolismo , Antioxidantes/metabolismo , Disponibilidade Biológica , Transporte Biológico
3.
Molecules ; 28(3)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36771156

RESUMO

PURPOSE: Propofol is a relatively short-acting potent anesthetic lipophilic drug used during short surgical procedures. Despite the success of propofol intravenous emulsions, drawbacks to such formulations include inherent emulsion instability, the lack of a safe vehicle to prevent sepsis, and concern regarding hyperlipidemia-related side effects. The aim of the current investigation was to develop a novel, lipid-based self-nanoemulsifying drug delivery system (SNEDDS) for propofol with improved stability and anesthetic activity for human use. METHODS: A series of SNEDDS formulations were developed using naturally obtained medium-chain/long-chain mono-, di-, and triglycerides, glyceryl monocaprylate, and water-soluble cosolvents with hydrogenated castor oil constructing ternary phase diagrams for propofol. The developed SNEDDS formulations were characterized using visual observation, particle size analysis, zeta potential, transmission electron microscopy, equilibrium solubility, in vitro dynamic dispersion and stability, and in vivo sleeping disorder studies in rats. The in vivo bioavailability of the SNEDDSs in rats was also studied to compare the representative formulations with the marketed product Diprivan®. RESULTS: Medium-chain triglycerides (M810) with mono-diglycerides (CMCM) as an oil blend and hydrogenated castor oil (KHS15) as a surfactant were selected as key ingredients in ternary phase diagram studies. The nanoemulsifying regions were identified from the studies and a number of SNEDDSs were formulated. Results from the characterization studies demonstrated the formation of efficient nanosized particles (28-45 nm globule size, 0.10-0.20 PDI) in the optimized SNEDDS with a drug loading of 50 mg/g, which is almost 500-fold higher than free propofol. TEM analysis showed the formation of spherical and homogeneous nanoparticles of less than 50 nm. The dissolution rate of the representative SNEDDS was faster than raw propofol and able to maintain 99% propofol in aqueous solution for around 24 h. The optimized liquid SNEDDS formulation was found to be thermodynamically stable. The intravenous administration of the SNEDDS in male Wistar rats induced a sleeping time of 73-88 min. The mean plasma concentrations after the IV administration of propofol nano-formulations PF2-SNEDDS and PF8-SNEDDS were 1348.07 ± 27.31 and 1138.66 ± 44.97 µg/mL, as compared to 891.44 ± 26.05 µg/mL (p = 0.05) observed after the IV administration of raw propofol. CONCLUSION: Propofol-loaded SNEDDS formulations could be a potential pharmaceutical product with improved stability, bioavailability, and anesthetic activity.


Assuntos
Nanopartículas , Propofol , Ratos , Masculino , Humanos , Animais , Ratos Wistar , Óleo de Rícino , Sistemas de Liberação de Medicamentos/métodos , Solubilidade , Emulsões , Disponibilidade Biológica , Triglicerídeos , Administração Intravenosa , Tamanho da Partícula , Administração Oral , Liberação Controlada de Fármacos
4.
Mol Ther ; 28(5): 1263-1275, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32145202

RESUMO

Tumor-targeting oncolytic viruses such as vaccinia virus (VV) are attractive cancer therapeutic agents that act through multiple mechanisms to provoke both tumor lysis and anti-tumor immune responses. However, delivery of these agents remains restricted to intra-tumoral administration, which prevents effective targeting of inaccessible and disseminated tumor cells. In the present study we have identified transient pharmacological inhibition of the leukocyte-enriched phosphoinositide 3-kinase δ (PI3Kδ) as a novel mechanism to potentiate intravenous delivery of oncolytic VV to tumors. Pre-treatment of immunocompetent mice with the PI3Kδ-selective inhibitor IC87114 or the clinically approved idelalisib (CAL-101), prior to intravenous delivery of a tumor-tropic VV, dramatically improved viral delivery to tumors. This occurred via an inhibition of viral attachment to, but not internalization by, systemic macrophages through perturbation of signaling pathways involving RhoA/ROCK, AKT, and Rac. Pre-treatment using PI3Kδ-selective inhibitors prior to intravenous delivery of VV resulted in enhanced anti-tumor efficacy and significantly prolonged survival compared to delivery without PI3Kδ inhibition. These results indicate that effective intravenous delivery of oncolytic VV may be clinically achievable and could be useful in improving anti-tumor efficacy of oncolytic virotherapy.


Assuntos
Adenina/análogos & derivados , Administração Intravenosa/métodos , Antineoplásicos/uso terapêutico , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Imunoterapia/métodos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Purinas/uso terapêutico , Quinazolinas/uso terapêutico , Quinazolinonas/uso terapêutico , Vaccinia virus/imunologia , Adenina/farmacologia , Adenina/uso terapêutico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Terapia Combinada/métodos , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Purinas/farmacologia , Quinazolinas/farmacologia , Quinazolinonas/farmacologia , Transplante Homólogo , Resultado do Tratamento , Carga Tumoral
5.
Mol Ther ; 28(10): 2150-2160, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32592687

RESUMO

The GM2 gangliosidoses, Tay-Sachs disease (TSD) and Sandhoff disease (SD), are fatal lysosomal storage disorders caused by mutations in the HEXA and HEXB genes, respectively. These mutations cause dysfunction of the lysosomal enzyme ß-N-acetylhexosaminidase A (HexA) and accumulation of GM2 ganglioside (GM2) with ensuing neurodegeneration, and death by 5 years of age. Until recently, the most successful therapy was achieved by intracranial co-delivery of monocistronic adeno-associated viral (AAV) vectors encoding Hex alpha and beta-subunits in animal models of SD. The blood-brain barrier crossing properties of AAV9 enables systemic gene therapy; however, the requirement of co-delivery of two monocistronic AAV vectors to overexpress the heterodimeric HexA protein has prevented the use of this approach. To address this need, we developed multiple AAV constructs encoding simultaneously HEXA and HEXB using AAV9 and AAV-PHP.B and tested their therapeutic efficacy in 4- to 6-week-old SD mice after systemic administration. Survival and biochemical outcomes revealed superiority of the AAV vector design using a bidirectional CBA promoter with equivalent dose-dependent outcomes for both capsids. AAV-treated mice performed normally in tests of motor function, CNS GM2 ganglioside levels were significantly reduced, and survival increased by >4-fold with some animals surviving past 2 years of age.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Doença de Sandhoff/terapia , Animais , Gerenciamento Clínico , Modelos Animais de Doenças , Gangliosídeo G(M2)/metabolismo , Expressão Gênica , Predisposição Genética para Doença , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Camundongos , Mutação , Doença de Sandhoff/genética , Doença de Tay-Sachs/genética , Doença de Tay-Sachs/metabolismo , Doença de Tay-Sachs/terapia , Transgenes , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
6.
Drug Dev Ind Pharm ; 41(9): 1416-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25204767

RESUMO

Niclosamide is an anthelmintic drug that also demonstrates great potential in fighting against cancers. However, parenteral delivery of niclosamide is challenged due to its insoluble property. This study aimed to develop an injectable formulation for niclosamide using nanocrystals. Niclosamide nanocrystals were prepared by wet media milling technique and characterized by electronic microscopes, differential scanning calorimetry, powder X-ray diffractometry and drug release, etc. The resulting nanocrystals using Tween 80 as the stabilizer were approximately 235 nm in particle size and showed a satisfactory stability. Pharmacokinetic studies revealed that there was no significant difference in plasma concentration-time profiles between nanocrystals and the control formulation (i.e. drug solution). By contrast, a significant difference in tissue distribution was observed at 2 h. Further, niclosamide nanocrystals presented a comparable antitumor effect to the drug solution against EC9076 cell line. We concluded that the nanocrystal formulation with solution-like behaviors should be a promising choice for intravenous delivery of niclosamide.


Assuntos
Anti-Helmínticos/administração & dosagem , Antineoplásicos/administração & dosagem , Nanopartículas , Niclosamida/administração & dosagem , Animais , Anti-Helmínticos/química , Anti-Helmínticos/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Varredura Diferencial de Calorimetria , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Humanos , Niclosamida/química , Niclosamida/farmacologia , Tamanho da Partícula , Polissorbatos/química , Ratos , Ratos Sprague-Dawley , Solubilidade , Fatores de Tempo , Distribuição Tecidual
7.
Int J Nanomedicine ; 19: 5707-5718, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38882540

RESUMO

Background: Rheumatoid Arthritis (RA) involves prolonged inflammation of the synovium, damaging joints and causing stiffness and deformity. Celastrol (Cel), derived from the Chinese herbal medicine Tripterygium wilfordii Hook F, offers immunosuppressive effects for RA treatment but is limited by poor solubility and bioavailability. Purpose: In this study, long-circulating Cel-loaded liposomes (Cel-LPs) were used to increase the pharmacokinetics of Cel, thereby improving drug delivery and efficacy for the treatment of RA. Methods: Cel-LPs were prepared and administered orally and intravenously to compare the elimination half-life of drugs and bioavailability of Cel. Cel-LPs were prepared using the lipid thin-layer-hydration-extrusion method. Human rheumatoid arthritis synovial (MH7A) cells were used to investigate the compatibility of Cel-LPs. The pharmacokinetic studies were performed on male Sprague-Dawley (SD) rats. Results: The Cel-LPs had an average size of 72.20 ± 27.99 nm, a PDI of 0.267, a zeta potential of -31.60 ± 6.81 mV, 78.77 ± 5.69% drug entrapment efficiency and sustained release (5.83 ± 0.42% drug loading). The cytotoxicity test showed that liposomes had excellent biocompatibility and the fluorescence microscope diagram indicated that liposome entrapment increased intracellular accumulation of Rhodamine B by MH7A cells. Furthermore, the results exhibited that Cel-LPs improved the pharmacokinetics of Cel by increasing the elimination half-life (t1/2) to 11.71 hr, mean residence time (MRT(0-∞)) to 7.98 hr and apparent volume of distribution (Vz/F) to 44.63 L/kg in rats, compared to the Cel solution. Conclusion: In this study, liposomes were demonstrated to be effective in optimizing the delivery of Cel, enabling the formulation of Cel-LPs with prolonged blood circulation and sustained release characteristics. This formulation enhanced the intravenous solubility and bioavailability of Cel, developing a foundation for its clinical application in RA and providing insights on poorly soluble drug management.


Assuntos
Lipossomos , Triterpenos Pentacíclicos , Ratos Sprague-Dawley , Triterpenos , Triterpenos Pentacíclicos/farmacocinética , Triterpenos Pentacíclicos/administração & dosagem , Animais , Lipossomos/química , Lipossomos/farmacocinética , Triterpenos/farmacocinética , Triterpenos/química , Triterpenos/administração & dosagem , Masculino , Humanos , Administração Intravenosa , Ratos , Disponibilidade Biológica , Linhagem Celular , Artrite Reumatoide/tratamento farmacológico , Tamanho da Partícula , Sobrevivência Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos
8.
ACS Nano ; 17(15): 14461-14474, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37367941

RESUMO

Intravenous administration of oncolytic adenoviruses (OVs) is a hopeful tumor therapeutic modality. However, the sharp clearance of OVs by the immune system dampens its effectiveness. Many studies have attempted to extend the circulation of intravenously administered OVs, almost all by preventing OVs from binding to neutralizing antibodies and complements in the blood, but the results have not been satisfactory. In contrast to previous conclusions, we found that the key to improving the circulation of OVs is to prevent the formation of the virus-protein corona rather than simply preventing the binding of neutralizing antibodies or complements to OVs. After identifying the key protein components of the virus-protein corona, we proposed a virus-protein corona replacement strategy, where an artificial virus-protein corona was formed on OVs to completely prevent the interaction of OVs with key virus-protein corona components in the plasma. It was found that this strategy dramatically prolonged the circulation time of OVs by over 30 fold and increased the distribution of OVs in tumors by over 10-fold, resulting in superior antitumor efficacy in primary and metastatic tumor models. Our finding provides a perspective on intravenous delivery of OVs, shifting the focus of future studies from preventing OV binding with neutralization antibodies and complements to preventing OVs from interacting with key virus-protein corona components in the plasma.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Coroa de Proteína , Humanos , Vírus Oncolíticos/genética , Terapia Viral Oncolítica/métodos , Adenoviridae/genética , Neoplasias/terapia , Anticorpos Neutralizantes
9.
Asian J Pharm Sci ; 18(1): 100771, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36896445

RESUMO

Current treatments for advanced hepatocellular carcinoma (HCC) have limited success in improving patients' quality of life and prolonging life expectancy. The clinical need for more efficient and safe therapies has contributed to the exploration of emerging strategies. Recently, there has been increased interest in oncolytic viruses (OVs) as a therapeutic modality for HCC. OVs undergo selective replication in cancerous tissues and kill tumor cells. Strikingly, pexastimogene devacirepvec (Pexa-Vec) was granted an orphan drug status in HCC by the U.S. Food and Drug Administration (FDA) in 2013. Meanwhile, dozens of OVs are being tested in HCC-directed clinical and preclinical trials. In this review, the pathogenesis and current therapies of HCC are outlined. Next, we summarize multiple OVs as single therapeutic agents for the treatment of HCC, which have demonstrated certain efficacy and low toxicity. Emerging carrier cell-, bioengineered cell mimetic- or nonbiological vehicle-mediated OV intravenous delivery systems in HCC therapy are described. In addition, we highlight the combination treatments between oncolytic virotherapy and other modalities. Finally, the clinical challenges and prospects of OV-based biotherapy are discussed, with the aim of continuing to develop a fascinating approach in HCC patients.

10.
Mol Ther Oncolytics ; 28: 334-348, 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36938544

RESUMO

VG2025 is a recombinant oncolytic herpes simplex virus type 1 (HSV-1) that uses transcriptional and translational dual regulation (TTDR) of critical viral genes to enhance virus safety and promote tumor-specific virus replication without reducing virulence. The TTDR platform is based on transcriptional control of the essential HSV-1 immediate-early protein ICP27 using a tumor-specific carcinoembryonic antigen (CEA) promoter, coupled with translational control of the neurovirulence factor ICP34.5 using multiple microRNA (miR)-binding sites. VG2025 further incorporates IL-12 and the IL-15/IL-15 receptor alpha subunit complex to enhance the antitumor and immune stimulatory properties of oncolytic HSVs. The TTDR strategy was verified in vitro and shown to be highly selective. Strong in vivo antitumor efficacy was observed following both intratumoral and intravenous administration. Clear abscopal and immune memory effects were also evident, indicating a robust antitumor immune response. Gene expression profiling of treated tumors revealed increased immune cell infiltration and activation of multiple immune-signaling pathways when compared with the backbone virus. Absence of neurotoxicity was verified in mice and in rhesus monkeys. Taken together, the enhanced tumor clearance, excellent safety profile, and positive correlation between CEA levels and viral replication efficiency may provide an opportunity for using biomarker-based precision medicine in oncolytic virotherapy.

11.
JMIR Res Protoc ; 11(7): e37898, 2022 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-35793128

RESUMO

BACKGROUND: Brain injuries resulting from motor vehicle accidents and falls, as well as hypoxic insults and other conditions, are one of the leading causes of disability and death in the world. Current treatments are limited but include continuous rehabilitation, especially for chronic brain injury. Recent studies have demonstrated that the intravenous infusion of mesenchymal stem cells (MSCs) has therapeutic efficacy for several neurological diseases, including stroke and spinal cord injury. OBJECTIVE: The objective of our investigator-initiated clinical trial is to assess the safety and potential efficacy of the intravenous infusion of autoserum-expanded autologous MSCs for patients with chronic brain injury. METHODS: The (phase 2) trial will be a single-arm, open-label trial with the primary objective of confirming the safety and efficacy of autoserum-expanded autologous MSCs (STR-01; produced under good manufacturing practices) when administered to patients with chronic brain injury. The estimated number of enrolled participants is 6 to 20 patients with a modified Rankin Scale grade of 3 to 5. The assessment of safety and the proportion of cases in which the modified Rankin Scale grade improves by 1 point or more at 180 days after the injection of STR-01 will be performed after MSC infusion. RESULTS: We received approval for our clinical trial from the Japanese Pharmaceuticals and Medical Devices Agency on December 12, 2017. The trial will be completed on June 11, 2023. The registration term is 5 years. The recruitment of the patients for this trial started on April 20, 2018, at Sapporo Medical University Hospital in Japan. CONCLUSIONS: Our phase 2 study will aim to address the safety and efficacy of the intravenous infusion of MSCs for patients with chronic brain injury. The use of STR-01 has been performed for patients with cerebral infarction and spinal cord injury, providing encouraging results. The potential therapeutic efficacy of the systemic administration of autoserum-expanded autologous MSCs for chronic brain injury should be evaluated, given its safety and promising results for stroke and spinal cord injury. TRIAL REGISTRATION: Japan Medical Association Center for Clinical Trials JMA-IIA00333; https://tinyurl.com/nzkdfnbc. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/37898.

12.
Front Neurol ; 12: 685802, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34512509

RESUMO

Widespread transduction of the CNS with a single, non-invasive systemic injection of adeno-associated virus is now possible due to the creation of blood-brain barrier-permeable capsids. However, as these capsids are mutants of AAV9, they do not have specific neuronal tropism. Therefore, it is necessary to use genetic tools to restrict expression of the transgene to neuronal tissues. Here we compare the strength and specificity of two neuron-specific promoters, human synapsin 1 and mouse calmodulin/calcium dependent kinase II, to the ubiquitous CAG promoter. Administration of a high titer of virus is necessary for widespread CNS transduction. We observed the neuron-specific promoters drive comparable overall expression in the brain to the CAG promoter. Furthermore, the neuron-specific promoters confer significantly less transgene expression in peripheral tissues compared with the CAG promoter. Future experiments will utilize these delivery platforms to over-express the Alzheimer-associated pathological proteins amyloid-beta and tau to create mouse models without transgenesis.

13.
Int J Pharm ; 586: 119555, 2020 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-32562654

RESUMO

Intravenous (IV) route is preferred for rapid onset of action, avoiding first pass metabolism and achieving site specific delivery. Development of IV formulations for poorly water soluble drugs poses significant challenges. Formulation approaches like salt formation, co-solvents, surfactants and inclusion complexation using cyclodextrins are used for solubilisation. However, these approaches are not applicable universally and have limitations in extent of solubilisation, hypersensitivity, toxicity and application to only specific type of molecules. IV nanosuspension have been attracting attention as a viable strategy for development of IV formulations of poorly water-soluble drugs. Nanosuspension consists of nanocrystals of poorly water soluble drug suspended in aqueous media and stabilized using minimal concentration of stabilizers. Recent years have witnessed their potential in formulations for toxicological studies and clinical trials. However various challenges are associated with the translational development of IV nanosuspensions. Therefore, the objective of the current review is to provide a holistic view of formulation development and desired properties of IV nanosuspensions. It will also focus on advancements in characterization tools, manufacturing techniques and post-production processing. Challenges associated with translational development and regulatory aspects of IV nanosuspension will be addressed. Additionally, their role in preclinical evaluation and special applications like targeting will also be discussed with the help of case studies. The applications of IV nanosuspensions shall expand as their applications move from preclinical phase to commercialization.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas , Preparações Farmacêuticas/administração & dosagem , Administração Intravenosa , Animais , Química Farmacêutica , Humanos , Preparações Farmacêuticas/química , Solubilidade , Suspensões , Tecnologia Farmacêutica , Água/química
14.
Mol Genet Metab Rep ; 24: 100604, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32461912

RESUMO

Mucopolysaccharidosis type I (MPS I) is an inherited metabolic disorder caused by deficiency of alpha-L-iduronidase (IDUA), resulting in accumulation of heparan and dermatan sulfate glycosaminoglycans (GAGs). Individuals with the most severe form of the disease (Hurler syndrome) suffer from neurodegeneration, intellectual disability, and death by age 10. Current treatments for this disease include allogeneic hematopoietic stem cell transplantation (HSCT) and enzyme replacement therapy (ERT). However, these treatments do not address CNS manifestations of the disease. In this study we compared the ability of intravenously administered AAV serotypes 9 and rh10 (AAV9 and AAVrh10) for delivery and expression of the IDUA gene in the CNS. Adult C57BL/6 MPS I mice were infused intravenously with either AAV9 or AAVrh10 vector encoding the human IDUA gene. Treated animals demonstrated supraphysiological levels and widespread restoration of IDUA enzyme activity in the plasma and all organs including the CNS. High levels of IDUA enzyme activity were observed in the plasma, brain and spinal cord ranging from 10 to 100-fold higher than heterozygote controls, while levels in peripheral organs were also high, ranging from 1000 to 10,000-fold higher than control animals. In general, levels of IDUA expression were slightly higher in peripheral organs for AAVrh10 administered animals although these differences were not significant except for the lung. Levels of IDUA expression between AAV 9 and rh10 were roughly equivalent in the brain. Urinary and tissue GAGs were significantly reduced starting at 3 weeks after vector infusion, with restoration of normal GAG levels by the end of the study in animals treated with either AAV9 or rh10. These results demonstrate that non-invasive intravenous AAV9 or AAVrh10-mediated IDUA gene therapy is a potentially effective treatment for both systemic and CNS manifestations of MPS I, with implications for the treatment of other metabolic and neurological diseases as well.

15.
Thyroid ; 29(6): 830-844, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30929576

RESUMO

Background: Medullary thyroid carcinoma (MTC) presents a disproportionate number of thyroid cancer deaths due to limited treatment options beyond surgery. Gain-of-function mutations of the human REarranged during Transfection (RET) proto-oncogene have been well-established as the key driver of MTC tumorigenesis. RET has been targeted by tyrosine kinase inhibitors (TKIs), such as cabozantinib and vandetanib. However, clinical results have been disappointing, with regular dose reductions and inevitable progression. This study aimed to identify RET-regulated microRNAs (miRNAs) and explore their potential as novel therapeutic targets. Methods: Small RNA sequencing was performed in MTC TT cells before and after RET inhibition to identify RET-regulated miRNAs of significance. In vitro gain-of-function studies were performed to investigate cellular and molecular effects of potential miRNAs on cell phenotypes. Systemic delivery of miRNA in MTC xenografts using EDV™ nanocells, targeted to epidermal growth factor receptor on tumor cells, was employed to assess the therapeutic potential and possible modulation of TKI responses. Results: The study demonstrates the tumor suppressive role of a specific RET-regulated miRNA, microRNA-153-3p (miR-153-3p), in MTC. Targeted intravenous delivery of miR-153-3p impeded the tumor growth in MTC xenografts. Furthermore, combined treatment with miR-153-3p plus cabozantinib caused greater growth inhibition and appeared to reverse cabozantinib resistance. Mechanistically, miR-153-3p targets ribosomal protein S6 kinase B1 (RPS6KB1) of mTOR signaling and reduced downstream phosphorylation of Bcl-2 associated death promoter. Conclusion: This study provides evidence to establish systemic miRNA replacement plus TKIs as a novel therapeutic for patients with metastatic, progressive MTC.


Assuntos
Carcinoma Medular/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Anilidas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Medular/genética , Carcinoma Medular/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , MicroRNAs/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-ret/genética , Piridinas/farmacologia , Receptores Proteína Tirosina Quinases/farmacologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia
16.
Curr Med Chem ; 25(41): 5755-5771, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29473493

RESUMO

Alzheimer's and Parkinson's diseases are the most common neurodegenerative diseases worldwide and their incidence is increasing due to the aging population. At the moment, the available therapies are not disease modifying and have several limitations, some of which are discussed in this review. One of the main limitations of these treatments is the low concentration that drugs reach in the central nervous system after systemic administration. Indeed, the presence of biological barriers, particularly the blood-brain barrier (BBB), hinders the effective drug delivery to the brain, reducing the potential benefit coming from the administration of the medication. In this review, the mechanisms of transport across the BBB and new methods to improve drug passage across the BBB are discussed. These methods include non-invasive solutions such as intranasal and intravitreal administration, and the use of nanotechnology solutions based on polymeric carriers when the drug is intravenously injected, orally taken for intestine adsorption or delivered through the dermal mucosa. Also, it provides an analysis of more invasive solutions that include intracranially injected hydrogels and implanted devices for local drug delivery. Efforts in finding new therapeutic drugs blocking neurodegenerative disease progression or reverting their course should be coupled with efforts addressed to efficient drug delivery systems. Hence, new pharmacology discoveries together with advancements in nanotechnologies and biomaterials for regenerative medicine are required to effectively counteract neurodegenerative diseases.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Portadores de Fármacos/administração & dosagem , Doença de Parkinson/tratamento farmacológico , Polímeros/química , Animais , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo , Liberação Controlada de Fármacos , Humanos , Hidrogéis/química , Absorção Intestinal , Nanopartículas/química , Tamanho da Partícula , Permeabilidade , Propriedades de Superfície
17.
Front Oncol ; 7: 208, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28971063

RESUMO

Nanotechnology has opened up a new, previously unimaginable world in cancer diagnosis and therapy, leading to the emergence of cancer nanomedicine and nanoparticle-aided radiotherapy. Smart nanomaterials (nanoparticle drones) can now be constructed with capability to precisely target cancer cells and be remotely activated with radiation to emit micrometer-range missile-like electrons to destroy the tumor cells. These nanoparticle drones can also be programmed to deliver therapeutic payloads to tumor sites to achieve optimal therapeutic efficacy. In this article, we examine the state-of-the-art and potential of nanoparticle drones in targeting lung cancer. Inhalation (INH) (air) versus traditional intravenous ("sea") routes of navigating physiological barriers using such drones is assessed. Results and analysis suggest that INH route may offer more promise for targeting tumor cells with radiosensitizers and cannabinoids from the perspective of maximizing damage to lung tumors cells while minimizing any collateral damage or side effects.

18.
Neurobiol Aging ; 51: 83-96, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28056358

RESUMO

The impact of human adult ischemia-tolerant mesenchymal stem cells (hMSCs) and factors (stem cell factors) on cerebral amyloid beta (Aß) pathology was investigated in a mouse model of Alzheimer's disease (AD). To this end, hMSCs were administered intravenously to APPPS1 transgenic mice that normally develop cerebral Aß. Quantitative reverse transcriptase polymerase chain reaction biodistribution revealed that intravenously delivered hMSCs were readily detected in APPPS1 brains 1 hour following administration, and dropped to negligible levels after 1 week. Notably, intravenously injected hMSCs that migrated to the brain region were localized in the cerebrovasculature, but they also could be observed in the brain parenchyma particularly in the hippocampus, as revealed by immunohistochemistry. A single hMSC injection markedly reduced soluble cerebral Aß levels in APPPS1 mice after 1 week, although increasing several Aß-degrading enzymes and modulating a panel of cerebral cytokines, suggesting an amyloid-degrading and anti-inflammatory impact of hMSCs. Furthermore, 10 weeks of hMSC treatment significantly reduced cerebral Aß plaques and neuroinflammation in APPPS1 mice, without increasing cerebral amyloid angiopathy or microhemorrhages. Notably, a repeated intranasal delivery of soluble factors secreted by hMSCs in culture, in the absence of intravenous hMSC injection, was also sufficient to diminish cerebral amyloidosis in the mice. In conclusion, this preclinical study strongly underlines that cerebral amyloidosis is amenable to therapeutic intervention based on peripheral applications of hMSC or hMSC factors, paving the way for a novel therapy for Aß amyloidosis and associated pathologies observed in AD.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Terapia de Alvo Molecular , Animais , Movimento Celular , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Injeções Intravenosas , Camundongos Transgênicos
19.
Int J Nanomedicine ; 10: 2521-35, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25848269

RESUMO

INTRODUCTION: SNX-2112 is a promising anticancer agent but has poor solubility in both water and oil. In the study reported here, we aimed to develop a nanocrystal formulation for SNX-2112 and to determine the pharmacokinetic behaviors of the prepared nanocrystals. METHODS: Nanocrystals of SNX-2112 were prepared using the wet-media milling technique and characterized by particle size, differential scanning calorimetry, drug release, etc. Physiologically based pharmacokinetic (PBPK) modeling was undertaken to evaluate the drug's disposition in rats following administration of drug cosolvent or nanocrystals. RESULTS: The optimized SNX-2112 nanocrystals (with poloxamer 188 as the stabilizer) were 203 nm in size with a zeta potential of -11.6 mV. In addition, the nanocrystals showed a comparable release profile to the control (drug cosolvent). Further, the rat PBPK model incorporating the parameters of particulate uptake (into the liver and spleen) and of in vivo drug release was well fitted to the experimental data following administration of the drug nanocrystals. The results reveal that the nanocrystals rapidly released drug molecules in vivo, accounting for their cosolvent-like pharmacokinetic behaviors. Due to particulate uptake, drug accumulation in the liver and spleen was significant at the initial time points (within 1 hour). CONCLUSION: The nanocrystals should be a good choice for the systemic delivery of the poorly soluble drug SNX-2112. Also, our study contributes to an improved understanding of the in vivo fate of nanocrystals.


Assuntos
Antineoplásicos , Portadores de Fármacos/química , Compostos Heterocíclicos de 4 ou mais Anéis , Nanopartículas/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacocinética , Fígado/metabolismo , Ratos , Baço/metabolismo
20.
Drug Deliv ; 22(2): 223-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24559497

RESUMO

Tamibarotene (Am80), a poorly water-soluble drug for the treatment of acute promyelocytic leukemia (APL), loaded nanostructured lipid carrier (Am80-NLC) was developed and characterized previously. The purpose of the present work was to develop PEGylated nanostructured lipid carrier (PEG-NLC) for intravenous delivery of Am80, with the aim to further extend the circulation in blood and decrease the adverse events. Am80-loaded PEG-NLC (Am80-PEG-NLC) modified with PEG-40 stearate (PEG40-SA, molecular weight 2000 Da) was formulated by the method of melt-emulsification and low temperature-solidification technique. Am80-NLC was developed as well as control. Based on the optimized results of single-factor screening experiment, the average drug entrapment efficiency, the mean particle size, and zeta potential of Am80-NLC and Am80-PEG-NLC were found to be 89.8-94.3%, 178.9-201.6 nm, and -37.74 to -20.1 mV, respectively. In vitro drug release of Am80-NLC and Am80-PEG-NLC possessed a sustained release characteristic and their release behavior was in accordance with the Ritger-Peppas equation. In vivo, after intravenous (i.v.) injection to rats, the mean residence time (MRT) of Am80-PEG-NLC group was significantly prolonged and the AUC value was improved as well compared with the Am80-NLC group. Furthermore, the biodistribution in mice showed that Am80-PEG-NLC preferentially decreased the accumulation of Am80 in kidney and increased the drug concentration in brain after i.v. injection. In conclusion, Am80-PEG-NLC may be a potential delivery system for Am80 in the treatment of APL.


Assuntos
Antineoplásicos/administração & dosagem , Benzoatos/administração & dosagem , Sistemas de Liberação de Medicamentos , Lecitinas/química , Nanoestruturas/química , Polietilenoglicóis/química , Tetra-Hidronaftalenos/administração & dosagem , Animais , Animais não Endogâmicos , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Benzoatos/química , Benzoatos/metabolismo , Benzoatos/farmacocinética , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Composição de Medicamentos , Meia-Vida , Injeções Intravenosas , Rim/efeitos dos fármacos , Rim/metabolismo , Leucemia Promielocítica Aguda/tratamento farmacológico , Camundongos , Nanoestruturas/ultraestrutura , Tamanho da Partícula , Distribuição Aleatória , Ratos Wistar , Solubilidade , Propriedades de Superfície , Tetra-Hidronaftalenos/química , Tetra-Hidronaftalenos/metabolismo , Tetra-Hidronaftalenos/farmacocinética , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA