Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Pharm Res ; 33(9): 2218-2228, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27251414

RESUMEN

PURPOSE: To investigate the applicability of fusion biotoxins combining pore-forming toxins (PFTs) and ribosome-inactivating proteins (RIPs) for the anti-cancer treatment. METHODS: Membrane active PFTs tend to destabilize cell membranes of tumor cells, but lack a warhead inducing significant cause of cell death. Cell-impermeable RIPs possess a powerful warhead, yet not able to enter the tumor cells. To address these challenges for anti-tumor effects, we introduced a fusion strategy of conjugating melittin (a PFT) and gelonin (a type 1 RIP) via chemical and recombinant methods, followed by in vitro assays and in vivo animal studies. RESULTS: In vitro characterization results confirmed that the chimeric gelonin-melittin fusion proteins retained equivalent intrinsic activity to that of unmodified gelonin in inhibiting protein translation. However, chemically conjugated gelonin-melittin (cGel-Mel) and recombinant chimeric gelonin-melittin fusion (rGel-Mel) exhibited greater cell uptake, yielding a significantly enhanced cytotoxic activity over treatment of gelonin, melittin or physical mixture of gelonin and melittin. Remarkably, cGel-Mel and rGel-Mel displayed 32- and 10-fold lower IC50 than gelonin in the cell lines. The superior anti-tumor efficacy of multivalent cGel-Mel to monovalent rGel-Mel suggested that valency could be a crucial factor for the extent of melittin-mediated cell uptake. Tumoricidal effects observed from animal studies were in good accordance with our findings from the cellular assays. CONCLUSIONS: This study successfully demonstrated that fusion of biotoxins could provide a simple yet effective way to synergistically augment their anti-tumor activity.


Asunto(s)
Antineoplásicos Fitogénicos/química , Meliteno/química , Proteínas Recombinantes de Fusión/química , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Toxinas Biológicas/química , Animales , Antineoplásicos Fitogénicos/farmacología , Línea Celular , Línea Celular Tumoral , Perros , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Meliteno/farmacología , Ratones , Neoplasias/tratamiento farmacológico , Ratas , Proteínas Recombinantes de Fusión/farmacología , Proteínas Inactivadoras de Ribosomas/química , Proteínas Inactivadoras de Ribosomas/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Toxinas Biológicas/farmacología
2.
BMC Ophthalmol ; 16: 43, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27095023

RESUMEN

BACKGROUND: The aim of this study was to analyze the shifts in retinal vessel diameter and oxygen saturation in diabetic patients with and without diabetic retinopathy (DR), as well as to assess the association between diabetes duration and either vessel diameter or oxygen saturation. METHODS: In total, 99 Type 2 DM patients were recruited for the study and were divided into three groups: DM with non-obvious retinopathy (DM, n = 29), non-proliferative diabetic retinopathy (NPDR, n = 40), and proliferative diabetic retinopathy (PDR, n = 30). In addition, 78 age-matched healthy individuals were chosen as the control. The diameter and oxygen saturation of the retinal vessels were analyzed using a noninvasive retinal oximeter, and then compared between the three groups and the normal control. Association analysis was applied to analyze the possible influencing factors, including the diameter and oxygen saturation of retinal vessels, on best corrected visual acuity BCVA, as well as the relationship between diabetes duration and the oximetry values. RESULTS: All of the diabetic patients showed thinner arterioles, wider venules, and a smaller arteriolar-to-venular ratio (AVR) than the healthy individuals. The AVR results from the controls through to the PDR group were 0.81 ± 0.07, 0.78 ± 0.07, 0.76 ± 0.07 and 0.67 ± 0.07, respectively. Both the NPDR and PDR groups showed significantly smaller AVR than the control. All of the diabetic patients exhibited higher retinal vessel oxygen saturation than the healthy individuals. Among all of the oximetry values, AVR exhibited the most significant correlation with best corrected visual acuity (BCVA) (ß = 1.533, P < 0.0001). An increased diabetes duration was associated with decreased arteriolar diameter (slope = -0.082 pixels/year, r (2) = 0.085, P = 0.004) and AVR (slope = -0.009/year, r (2) = 0.349, P < 0.001), and with increased venular diameter (slope = 0.104 pixels/year, r (2) = -0.109, P = 0.001). CONCLUSIONS: In this Chinese population with type 2 DM, the thinner arterioles and wider venules point to microvascular dysfunction in DR. The increased oxygen saturation of the retinal vessels suggests that retinal oxygen metabolism is affected in diabetic retinopathy.


Asunto(s)
Diabetes Mellitus Tipo 2/diagnóstico , Retinopatía Diabética/diagnóstico , Oxígeno/sangre , Arteria Retiniana/patología , Vena Retiniana/patología , Anciano , Arteriolas/patología , Pueblo Asiatico/etnología , China/epidemiología , Diabetes Mellitus Tipo 2/etnología , Diabetes Mellitus Tipo 2/fisiopatología , Retinopatía Diabética/etnología , Retinopatía Diabética/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oximetría , Vénulas/patología , Agudeza Visual/fisiología
3.
Int J Mol Sci ; 17(11)2016 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-27854260

RESUMEN

For the past 20 years, we have witnessed an unprecedented and, indeed, rather miraculous event of how cell-penetrating peptides (CPPs), the naturally originated penetrating enhancers, help overcome the membrane barrier that has hindered the access of bio-macromolecular compounds such as genes and proteins into cells, thereby denying their clinical potential to become potent anti-cancer drugs. By taking the advantage of the unique cell-translocation property of these short peptides, various payloads of proteins, nucleic acids, or even nanoparticle-based carriers were delivered into all cell types with unparalleled efficiency. However, non-specific CPP-mediated cell penetration into normal tissues can lead to widespread organ distribution of the payloads, thereby reducing the therapeutic efficacy of the drug and at the same time increasing the drug-induced toxic effects. In view of these challenges, we present herein a review of the new designs of CPP-linked vehicles and strategies to achieve highly effective yet less toxic chemotherapy in combating tumor oncology.


Asunto(s)
Antineoplásicos/administración & dosificación , Péptidos de Penetración Celular/administración & dosificación , Portadores de Fármacos/administración & dosificación , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacocinética , Péptidos de Penetración Celular/farmacocinética , Portadores de Fármacos/farmacocinética , Endocitosis , Humanos , Concentración de Iones de Hidrógeno , Permeabilidad , Distribución Tisular
4.
Pharm Res ; 32(8): 2690-703, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25701313

RESUMEN

PURPOSE: To investigate the feasibility of applying PTD-modified ATTEMPTS (Antibody Targeted Triggered Electrically Modified Prodrug-Type Strategy) for enhanced toxin therapy for the treatment of cancer. METHODS: A heparin-functionalized murine anti-CEA monoclonal antibody (mAb), T84.66-heparin (T84.66-Hep), was chemically synthesized and characterized for specific binding to CEA overexpressed cells. The T84.66-Hep was then applied to the PTD-modified ATTEMPTS approach and the crucial features of the drug delivery system (DDS), 'antibody targeting' and 'heparin/protamine-based prodrug', were evaluated in vitro to examine whether it could selective delivery a PTD-modified toxin, recombinant TAT-gelonin chimera (TAT-Gel), to CEA high expression cancer cells (LS174T). Furthermore, the feasibility of the drug delivery system (DDS) was assessed in vivo by biodistribution and efficacy studies using LS174T s.c. xenograft tumor bearing mice. RESULTS: T84.66-Hep displayed specific binding, but limited internalization (35% after 48 h incubation) to CEA high expression LS174T cells over low expression HCT116 cells. When mixed together with TAT-Gel, the T84.66-Hep formed a strong yet reversible complex. This complex formation provided an effective means of active tumor targeting of TAT-Gel, by 1) directing the TAT-Gel to CEA overexpressed tumor cells and 2) preventing nonspecific cell transduction to non-targeted normal cells. The cell transduction of TAT-Gel could, however, be efficiently reversed by addition of protamine. Feasibility of in vivo tumor targeting and "protamine-induced release" of TAT-Gel from the T84.66-Hep counterpart was confirmed by biodistribution and preliminary efficacy studies. CONCLUSIONS: This study successfully demonstrated in vitro and in vivo the applicability of PTD-modified ATTEMPTS for toxin-based cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Profármacos/metabolismo , Animales , Antineoplásicos/farmacología , Antígeno Carcinoembrionario/metabolismo , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Heparina/administración & dosificación , Heparina/uso terapéutico , Humanos , Ratones , Ratones Desnudos , Proteínas Mutantes Quiméricas , Protaminas/administración & dosificación , Protaminas/uso terapéutico , Proteínas/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Transducción de Señal/genética , Transducción de Señal/fisiología , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Ophthalmic Physiol Opt ; 35(5): 562-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26303449

RESUMEN

PURPOSE: To investigate changes in retinal vessel oxygen saturation and diameter in high myopia. METHODS: Relative oxygen saturation was measured in the retinal blood vessels of 54 participants with high myopia and compared to a control group of 54 individuals with emmetropia with the Oxymap T1 retinal oximeter. The participants with high myopia were further divided into two groups according to the grade of myopic retinopathy: Group A (grade < M2 ) and Group B (grade ≥ M2 ). One-way anova was used to analyse the mean saturation and diameter of retinal arterioles and venules and the mean difference in arterio-venous saturation among the four groups. Further analysis of multiple comparisons was performed with the Bonferroni test. Linear regression was used to analyse the correlation of ocular perfusion pressure or best corrected visual acuity with other variables. RESULTS: For all of the high myopia patients, retinal arteriole saturation (92.3 ± 5.6%) and the difference in arterio-venous saturation (30.8 ± 5.0%) were significantly lower than in normal individuals (96.0 ± 5.8%, 35.4 ± 6.2%; p = 0.006, p < 0.001, respectively). In Group A, only the difference in arterio-venous saturation (31.0 ± 4.7%) was significantly lower than in the control group (p = 0.011). In Group B, retinal arteriole saturation (92.2 ± 5.3%) and the difference in arterio-venous saturation (30.7 ± 5.3%) were also lower than the control group (p = 0.02, p = 0.001, respectively). Both retinal arteriole diameter and retinal venule diameter were narrower than in participants with high myopia than the control group (p < 0.001). No statistically significant correlations were found between ocular perfusion pressure or best corrected visual acuity with any other variables. CONCLUSIONS: The study demonstrated decreased retinal arteriole saturation and decreased difference in arterio-venous saturation as well as narrowing retinal vessel diameter in highly myopic eyes. Further studies are needed to determine if such changes play a role in the development of high myopia and its complications or occur as a consequence of tissue remodelling during axial elongation.


Asunto(s)
Miopía/fisiopatología , Oxígeno/sangre , Vasos Retinianos/fisiopatología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Estudios de Casos y Controles , Niño , Femenino , Humanos , Presión Intraocular/fisiología , Masculino , Persona de Mediana Edad , Oximetría , Análisis de Regresión , Arteria Retiniana/fisiopatología , Vena Retiniana/fisiopatología , Adulto Joven
6.
ACS Nano ; 18(26): 17267-17281, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38871478

RESUMEN

Intrinsic or acquired resistance to chemical drugs severely limits their therapeutic efficacy in cancer treatment. Various intracellular antioxidant molecules, particularly glutathione (GSH), play a crucial role in maintaining intracellular redox homeostasis by mitigating the overproduced reactive oxygen species (ROS) due to rapid cell proliferation. Notably, these antioxidants also eliminate chemical-drug-induced ROS, eventually diminishing their cytotoxicity and rendering them less effective. In this study, we combined erastin, a GSH biosynthesis inhibitor, with 2'-deoxy-5-fluorouridine 5'-monophosphate sodium salt (FdUMP), an ROS-based drug, to effectively disrupt intracellular redox homeostasis and reverse chemotherapy resistance. Therefore, efficient ferroptosis and apoptosis were simultaneously induced for enhanced antitumor effects. Additionally, we employed small interfering RNA targeting PD-L1 (siPD-L1) as a third agent to block immune-checkpoint recognition by CD8+ T cells. The highly immunogenic cell peroxidates or damage-associated molecular patterns (DAMPs) induced by erastin acted synergistically with downregulated PD-L1 to enhance the antitumor effects. To codeliver these three drugs simultaneously and efficiently, we designed GE11 peptide-modified lipid nanoparticles (LNPs) containing calcium phosphate cores to achieve high encapsulation efficiencies. In vitro studies verified its enhanced cytotoxicity, efficient intracellular ROS induction and GSH/GPX4 downregulation, substantial lipid peroxidation product accumulation, and mitochondrial depolarization. In vivo, this formulation effectively accumulated at tumor sites and achieved significant tumor inhibition in subcutaneous colon cancer (CRC) mouse models with a maximum tumor inhibition rate of 83.89% at a relatively low dose. Overall, a strategy to overcome clinical drug resistance was verified in this study by depleting GSH and activating adaptive immunity.


Asunto(s)
Antineoplásicos , Apoptosis , Antígeno B7-H1 , Regulación hacia Abajo , Ferroptosis , Nanopartículas , Ferroptosis/efectos de los fármacos , Animales , Humanos , Ratones , Nanopartículas/química , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Regulación hacia Abajo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Lípidos/química , Proliferación Celular/efectos de los fármacos , Femenino , Ensayos de Selección de Medicamentos Antitumorales , Línea Celular Tumoral , Liposomas
7.
J Control Release ; 368: 52-65, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38368946

RESUMEN

FOLFOX regimen, composed of folinic acid, 5-fluorouracil (5-FU) and oxaliplatin (OXP), has been used as clinical standard therapeutic regimen in treatments of colorectal cancer (CRC) and esophageal squamous cell carcinoma (ESCC). To further improve its therapeutic outcomes, FOLFOX was combined with anti-PD-1 antibody to form an advanced chemo-immune combination strategy, which has been proven more efficient in controlling cancer progression and prolonging patients' survival in various clinical trials. However, bad tumor accumulation, relative high toxicity, numerous treatment cycles with high fees and low compliance as well as drug resistance seriously limit the prognosis of FOLFOX regimen. The "all-in-one" formulations, which could precisely delivery multidrug regimen into tumor sites and cells, showed a promising application prospect for targeted drug delivery as well as reducing side effects. However, the design and preparation of the "all-in-one" formulation with high drug encapsulation efficiencies for all drugs was still challenging. Herein, a lipid core-shell nanoparticle codelivery platform was designed for simultaneous encapsulation of variant FOLFOX composed of miriplatin (MiPt), 5-Fluoro-2'-deoxyuridine 5'-monophosphate (FdUMP), calcium folinate (CF) and PD-L1 siRNA (siPD-L1) with high efficiencies, and their synergistic anti-tumor mechanisms were studied, respectively. MiPt, a precursor of OXP, was validated capable of inducing efficient immunogenic cell death (ICD) in this work. Additionally, ICD-mediated release of damage associated molecular patterns functionalized synergistically with PD-L1 silence by siPD-L1 to overcome chemoresistance, reverse suppressive tumor microenvironment and recruit more CD8+ T cells. FdUMP, as the intracellular active form of 5-FU, could induce large amounts of reactive oxygen species to enhance the ICD. CF worked as the sensitizer of FdUMP. The enhanced long-term anti-tumor effect of the prepared "all-in-one" formulation compared to free drug regimen and other controls, was verified in heterotopic CRC mice models and ESCC mice models, providing new thoughts for researchers and showing a promising prospect of translation into clinical applications.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Colorrectales , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Nanopartículas , Humanos , Animales , Ratones , Leucovorina/uso terapéutico , Antígeno B7-H1 , Neoplasias Colorrectales/patología , Linfocitos T CD8-positivos/patología , Fluorodesoxiuridilato/uso terapéutico , Fluorouracilo/uso terapéutico , Oxaliplatino , Lípidos/uso terapéutico , Línea Celular Tumoral , Inmunoterapia , Compuestos Organoplatinos
8.
Mater Today Bio ; 26: 101016, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38516171

RESUMEN

Bone damage is a complex orthopedic problem primarily caused by trauma, cancer, or bacterial infection of bone tissue. Clinical care management for bone damage remains a significant clinical challenge and there is a growing need for more advanced bone therapy options. Nanotechnology has been widely explored in the field of orthopedic therapy for the treatment of a severe bone disease. Among nanomaterials, gold nanoparticles (GNPs) along with other biomaterials are emerging as a new paradigm for treatment with excellent potential for bone tissue engineering and regenerative medicine applications. In recent years, a great deal of research has focused on demonstrating the potential for GNPs to provide for enhancement of osteogenesis, reduction of osteoclastogenesis/osteomyelitis, and treatment of bone cancer. This review details the latest understandings in regards to GNPs based therapeutic systems, mechanisms, and the applications of GNPs against various bone disorders. The present review aims to summarize i) the mechanisms of GNPs in bone tissue remodeling, ii) preparation methods of GNPs, and iii) functionalization of GNPs and its decoration on biomaterials as a delivery vehicle in a specific bone tissue engineering for future clinical application.

9.
Mol Pharm ; 10(10): 3892-902, 2013 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-24024964

RESUMEN

Starch-coated, PEGylated, and heparin-functionalized iron oxide magnetic nanoparticles (DNPH) were successfully synthesized and characterized in detail. The PEGylation (20 kDa) process resulted in an average coating of 430 PEG molecules per nanoparticle. After that, heparin conjugation was carried out to attain the final DNPH platform with 35.4 µg of heparin/mg of Fe. Commercially acquired heparin-coated magnetic nanoparticles were also PEGylated (HP) and characterized for comparison. Protamine was selected as a model protein to demonstrate the strong binding affinity and high loading content of DNPH for therapeutically relevant cationic proteins. DNPH showed a maximum loading of 22.9 µg of protamine/mg of Fe. In the pharmacokinetic study, DNPH displayed a long-circulating half-life of 9.37 h, 37.5-fold longer than that (0.15 h) of HP. This improved plasma stability enabled extended exposure of DNPH to the tumor lesions, as was visually confirmed in a flank 9L-glioma mouse model using magnetic resonance imaging (MRI). Quantitative analysis of the Fe content in excised tumor lesions further demonstrated the superior tumor targeting ability of DNPH, with up to 31.36 µg of Fe/g of tissue (13.07% injected dose (I.D.)/g of tissue) and 7.5-fold improvement over that (4.27 µg of Fe/g of tissue; 1.78% I.D./g of tissue) of HP. Overall, this study shed light on the potential of DNPH to be used as a protein drug delivery platform.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Heparina/química , Imagen por Resonancia Magnética/métodos , Nanopartículas/química , Animales , Compuestos Férricos/química , Glioma/diagnóstico , Glioma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nanopartículas/metabolismo , Protaminas/química
10.
Pharm Res ; 30(10): 2445-58, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23344909

RESUMEN

In this review, we discussed the establishment of a so-called "theranostic" system by instituting the basic principles including the use of: [1] magnetic iron oxide nanoparticles (MION)-based drug carrier; [2] intra-arterial (I.A.) magnetic targeting; [3] macromolecular drugs with unmatched therapeutic potency and a repetitive reaction mechanism; [4] cell-penetrating peptide-mediated cellular drug uptake; and [5] heparin/protamine-regulated prodrug protection and tumor-specific drug re-activation into one single drug delivery system to overcome all possible obstacles, thereby achieving a potentially non-invasive, magnetic resonance imaging-guided, clinically enabled yet minimally toxic brain tumor drug therapy. By applying a topography-optimized I.A. magnetic targeting to dodge rapid organ clearance of the carrier during its first passage into the circulation, tumor capture of MION was enriched by >350 folds over that by conventional passive enhanced permeability and retention targeting. By adopting the prodrug strategy, we observed by far the first experimental success in a rat model of delivering micro-gram quantity of the large ß-galactosidase model protein selectively into a brain tumor but not to the ipsi- or contra-lateral normal brain regions. With the therapeutic regimens of most toxin/siRNA drugs to fully (>99.9%) eradicate a tumor being in the nano-molar range, the prospects of reaching this threshold become practically accomplishable.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Sistemas de Liberación de Medicamentos/métodos , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita , Animales , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Humanos , Inyecciones Intraarteriales , Inyecciones Intravenosas , Nanopartículas de Magnetita/administración & dosificación , Nanopartículas de Magnetita/uso terapéutico
11.
Acta Pharm Sin B ; 13(9): 3945-3955, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37719367

RESUMEN

Immunotherapy has revolutionized the landscape of cancer treatment. However, single immunotherapy only works well in a small subset of patients. Combined immunotherapy with antitumor synergism holds considerable potential to boost the therapeutic outcome. Nevertheless, the synergistic, additive or antagonistic antitumor effects of combined immunotherapies have been rarely explored. Herein, we established a novel combined cancer treatment modality by synergizing p21-activated kinase 4 (PAK4) silencing with immunogenic phototherapy in engineered extracellular vesicles (EVs) that were fabricated by coating M1 macrophage-derived EVs on the surface of the nano-complex cores assembled with siRNA against PAK4 and a photoactivatable polyethyleneimine. The engineered EVs induced potent PAK4 silencing and robust immunogenic phototherapy, thus contributing to effective antitumor effects in vitro and in vivo. Moreover, the antitumor synergism of the combined treatment was quantitatively determined by the CompuSyn method. The combination index (CI) and isobologram results confirmed that there was an antitumor synergism for the combined treatment. Furthermore, the dose reduction index (DRI) showed favorable dose reduction, revealing lower toxicity and higher biocompatibility of the engineered EVs. Collectively, the study presents a synergistically potentiated cancer treatment modality by combining PAK4 silencing with immunogenic phototherapy in engineered EVs, which is promising for boosting the therapeutic outcome of cancer immunotherapy.

12.
Acta Pharm Sin B ; 13(12): 5091-5106, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38045037

RESUMEN

Despite exciting achievements with some malignancies, immunotherapy for hypoimmunogenic cancers, especially glioblastoma (GBM), remains a formidable clinical challenge. Poor immunogenicity and deficient immune infiltrates are two major limitations to an effective cancer-specific immune response. Herein, we propose that an injectable signal-amplifying nanocomposite/hydrogel system consisting of granulocyte-macrophage colony-stimulating factor and imiquimod-loaded antigen-capturing nanoparticles can simultaneously amplify the chemotactic signal of antigen-presenting cells and the "danger" signal of GBM. We demonstrated the feasibility of this strategy in two scenarios of GBM. In the first scenario, we showed that this simultaneous amplification system, in conjunction with local chemotherapy, enhanced both the immunogenicity and immune infiltrates in a recurrent GBM model; thus, ultimately making a cold GBM hot and suppressing postoperative relapse. Encouraged by excellent efficacy, we further exploited this signal-amplifying system to improve the efficiency of vaccine lysate in the treatment of refractory multiple GBM, a disease with limited clinical treatment options. In general, this biomaterial-based immune signal amplification system represents a unique approach to restore GBM-specific immunity and may provide a beneficial preliminary treatment for other clinically refractory malignancies.

13.
J Control Release ; 345: 314-333, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35331783

RESUMEN

Since its outbreak in late 2019, the novel coronavirus disease 2019 (COVID-19) has spread to every continent on the planet. The global pandemic has affected human health and socioeconomic status around the world. At first, the global response to the pandemic was to isolate afflicted individuals to prevent the virus from spreading, while vaccine development was ongoing. The genome sequence was first presented in early January 2020, and the phase I clinical trial of the vaccine started in March 2020 in the United States using novel lipid-based nanoparticle (LNP), encapsulated with mRNA termed as mRNA-1273. Till now, various mRNA-based vaccines are in development, while one mRNA-based vaccine got market approval from US-FDA for the prevention of COVID-19. Previously, mRNA-based vaccines were thought to be difficult to develop, but the current development is a significant accomplishment. However, widespread production and global availability of mRNA-based vaccinations to combat the COVID-19 pandemic remains a major challenge, especially when the mutations continually occur on the virus (e.g., the recent outbreaks of Omicron variant). This review elaborately discusses the COVID-19 pandemic, the biology of SARS-CoV-2 and the progress of mRNA-based vaccines. Moreover, the review also highlighted a detailed description of mRNA delivery technologies and the application potential in controlling other life-threatening diseases. Therefore, it provides a comprehensive view and multidisciplinary insights into mRNA therapy for broader audiences.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Pandemias/prevención & control , ARN Mensajero/genética , SARS-CoV-2/genética , Estados Unidos/epidemiología , Vacunas Sintéticas , Vacunas de ARNm
14.
J Control Release ; 349: 565-579, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35835399

RESUMEN

Local treatment after resection to inhibit glioma recurrence is thought to able to meet the real medical needs. However, the only clinically approved local glioma treatment-wafer containing bis(2-chloroethyl) nitrosourea (BCNU) showed very limited effects. Herein, in order to inhibit tumor recurrence with prolonged and synergistic therapeutic effect of drugs after tumor resection, an in situ dual-sensitive hydrogel drug delivery system loaded with two synergistic chemo-drugs BCNU and temozolomide (TMZ) was developed. The thermosensitive hydrogel was loaded with reactive oxygen species (ROS)-sensitive poly (lactic-co-glycolic) acid nanoparticles (NPs) encapsulating both BCNU and TMZ and also free BCNU and TMZ. The in vitro synergistic effect of BCNU and TMZ and in vivo presence of ROS at the residual tumor site were confirmed. The prepared ROS-sensitive NPs and thermosensitive hydrogel, as well as the long-term release behavior of drugs and NPs, were fully characterized both in vitro and in vivo. After >90% glioblastoma resection, the dual-sensitive hydrogel drug delivery system was injected into the resection cavity. The median survival time of the experimental group reached 65 days which was twice as long as the Resection only group, implying that this in situ drug delivery system effectively inhibited tumor recurrence. Overall, this study provides new ideas and strategies for the inhibition of postoperative glioma recurrence.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Carmustina/uso terapéutico , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Glioma/patología , Glioma/cirugía , Humanos , Hidrogeles/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Especies Reactivas de Oxígeno , Temozolomida
15.
Biosaf Health ; 4(2): 70-78, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35310559

RESUMEN

Despite multiple virus outbreaks over the past decade, including the devastating coronavirus disease 2019 (COVID-19) pandemic, the lack of accurate and timely diagnosis and treatment technologies has wreaked havoc on global biosecurity. The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated proteins (Cas) system has the potential to address these critical needs for tackling infectious diseases to detect viral nucleic acids and inhibit viral replication. This review summarizes how the CRISPR/Cas system is being utilized for the treatment and diagnosis of infectious diseases with the help of biosafety materials and highlights the design principle and in vivo and in vitro efficacy of advanced biosafety materials used to deal with virus attacks.

16.
Int J Pharm ; 606: 120940, 2021 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-34310959

RESUMEN

The tissue-specific targeted delivery and efficient cellular uptake of siRNAs are the main obstacles to their clinical application. Antibody-siRNA-conjugates (ARCs) can deliver siRNA by exploiting the targeting property of antibodies like antibody-drug conjugates (ADCs). However, the effective conjugation of antibodies and siRNAs and the release of siRNAs specifically at target sites have posed challenges to the development of ARCs. In this study, the successful conjugation of antibodies and siRNAs was achieved using a multifunctional peptide as a linker, composed of a cell-penetrating peptide (CPP) and a substrate peptide (SP), which is highly expressed in solid tumors. The resulting antibody-multifunctional peptide (SP-CPP)-siRNA system delivered the siRNA to target tumor cells by the specific binding of the antibody. Once the enzymes on the tumor cell surface hydrolyzed the substrate peptide linker, siRNA-CPP was released from ARCs. The released siRNA-CPP entered the targeted cells via the cellular penetrating ability of CPP, resulting in improved siRNA-mediated gene silencing efficiency, verified both in vitro and in vivo. After intravenous administration, the designed ARCs achieved approximately 66.7% EGFP (Enhanced Green Fluorescent Protein) downregulation efficiency in nude mice xenografted with the HCT116-EGFP tumor model. The proposed system provides a prospective choice for ARC production and the safe and efficient delivery of siRNAs.


Asunto(s)
Péptidos de Penetración Celular , Inmunoconjugados , Animales , Línea Celular Tumoral , Ratones , Ratones Desnudos , Estudios Prospectivos , ARN Interferente Pequeño
17.
J Control Release ; 322: 610-621, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32194175

RESUMEN

Critical limb ischemia (CLI) is the most advanced stage of peripheral artery disease, associated with significant risk of limb loss, morbidity and mortality; however, there remains unmet therapeutic needs for arterial revascularization and ischemic tissue repair. Stem cell therapies have emerged as compelling candidates due to beneficial proangiogenic and immunosuppressive function. Nevertheless, in vivo efficacy was insufficient in proliferation, differentiation and survival/engraftment rate. Cardiac stem cells (CSCs) was firstly attempted for CLI as a novel therapeutic modality to provide superior angiogenic potency to bone marrow-derived stem cells (BMSCs). It was noted that CSCs demonstrated 3.2-fold in HGF, 2.9-fold in VEGF and 8.7-fold in PDGF-B higher gene expressions compared to BMSCs. To enhance the hypoxia-induced proangiogenic effect, CSCs were transfected with hypoxia-inducible factor-1 alpha (HIF-1α) by using electroporation method, specifically optimized for CSCs yielding 45.77% of transfection efficiency and 89.75% of viability. HIF-1α overexpression significantly increased CSC survival in hypoxia, proangiogenic factors production and endothelial differentiation. In mouse hind limb ischemia model, local intramuscular delivery of CSC overexpressing HIF-1α (HIF-CSC) significantly improved the blood flow recovery. Histological analysis revealed that muscle degeneration and fibrosis in the ischemic limb were attenuated. Local delivery of HIF-CSC might be a promising option for ischemic tissue restoration.


Asunto(s)
Células Madre Mesenquimatosas , Enfermedad Arterial Periférica , Animales , Diferenciación Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Isquemia/terapia , Ratones , Neovascularización Patológica , Neovascularización Fisiológica , Enfermedad Arterial Periférica/terapia
18.
Biomater Sci ; 8(8): 2308-2317, 2020 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-32186291

RESUMEN

Overlapping substrate specificities within the family of matrix metalloproteinases (MMPs), usually caused by their highly conserved structural topology, increase the potential for a substrate to be cleaved by multiple enzymes within this family, which leads to the decrease in the selectivity of MMP substrate-based probes. To resolve this issue, MT1-MMP activatable fluorogenic probes for tumor detection with enhanced specificity were developed by combining a fluorescence resonance energy transfer (FRET) peptide substrate and its specific binding peptide with different lengths of linkers. The specificity of the probes increased profiting from the high affinity of the MT1-MMP specific binding peptide while keeping the ability to amplify the output imaging signals in response to MMP activity with the FRET substrate. Enzyme kinetics analysis clearly demonstrated that the conjugation of P-1 and MT1-AF7p enhanced both the specificity and selectivity of the fluorogenic probes for MT1-MMP, and introducing a linker composed of 12 PEG subunits into these two fragments led to optimized specificity and selectivity of the fluorogenic probe for MT1-MMP. Both in vitro and in vivo results revealed that the imaging probe with the linker composed of 12 PEG subunits based on our designed strategy could be effectively applied for MT1-MMP positive tumor imaging. Since this strategy for enhancing the specificity of protease sensing probes can be applied to other proteases and is not just limited to MT1-MMP, it is an appealing platform to achieve selective tumor imaging.


Asunto(s)
Colorantes Fluorescentes/administración & dosificación , Metaloproteinasa 14 de la Matriz/administración & dosificación , Péptidos/administración & dosificación , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/química , Humanos , Metaloproteinasa 14 de la Matriz/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Péptidos/química , Proteínas Recombinantes/administración & dosificación
19.
Int J Pharm ; 584: 119469, 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32470482

RESUMEN

Non-injectable delivery of peptides and proteins is not feasible due to the limitations of large molecular mass, high hydrophilic properties, and gastrointestinal degradation. Therefore, proposing a new method to solve this problem is a burning issue. The objective of this study was to propose a novel protein delivery strategy to overcome the poor efficacy and irritation of buccal insulin delivery. In this study, we applied a conjugate of cell-penetrating peptides (LMWP) and insulin (INS-PEG-LMWP) for buccal delivery. INS-PEG-LMWP was prepared using insulin solution and mixture as references. The transport behaviour, in vivo bioactivity, hypoglycaemic effect, and safety of INS-PEG-LMWP were systematically characterised. An in vitro study demonstrated that the uptake and transportation of INS-PEG-LMWP across buccal mucosal multilayers significantly increased. By comparing the effects of different endocytic inhibitors on INS-PEG-LMWP uptake, the conjugate might be delivered via an energy independent, electrostatically adsorbed pathway. INS-PEG-LMWP's relative pharmacological bioavailability was high and its relative bioavailability was up to 26.86%, demonstrating no visible mucosal irritation. Cell-penetrating peptides are likely to become a reliable and safe tool for overcoming insulin's low permeability through the epithelial multilayers, the major barrier to buccal delivery.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Mucosa Bucal/metabolismo , Polietilenglicoles/administración & dosificación , Animales , Línea Celular Tumoral , Péptidos de Penetración Celular/farmacocinética , Humanos , Hipoglucemiantes/sangre , Hipoglucemiantes/farmacocinética , Insulina/sangre , Insulina/farmacocinética , Masculino , Absorción por la Mucosa Oral , Permeabilidad , Polietilenglicoles/farmacocinética , Conejos , Porcinos
20.
J Biomed Nanotechnol ; 15(3): 487-499, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-31165694

RESUMEN

Integrating the functions of bioimaging, targeting and controlled release of therapeutic agents into a single nanoparticle is of great interests in nanomedicine and nanobiology. Herein, a cis -diol/pH dual-responsive upconversion nanoparticle (UCNP)-based theranostic platform has been developed for delivery of the anticancer drug to cancer cells. This nanoplatform is based on the strategic design of targetable hyaluronan modified UCNPs (HA-UCNPs) that are coupled with aminobenzeneboronic acid (APBA) to obtain APBA-UCNPs, having favorable tumor selectivity as well as the capacity for capturing cis-diol-containing therapeutics. The controlled release function is then achieved through the self-assembly of hydroxycamptothecin derivative ligands onto the surfaces of APBA-UCNPs, which is controllable in a stimuli-dependent manner. The UCNP-based theranostic probe taken up by tumor cells via receptor-mediated endocytosis liberates drugs triggered by competitive glucose at low pH in endosomes/lysosomes, resulting in cell apoptosis. The dual-responsive mechanism of boronate ester bonds gives a chemoselective strategy for controlled release of drug within tumor cells, establishing an alternative approach to treat a broad spectrum of diseases exploiting similar boronic acid-involved therapeutics.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Ácido Hialurónico , Concentración de Iones de Hidrógeno , Nanomedicina
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda