Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 431
Filtrar
1.
Mol Ther ; 32(5): 1266-1283, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38569556

RESUMEN

Carrier-free naked mRNA vaccines may reduce the reactogenicity associated with delivery carriers; however, their effectiveness against infectious diseases has been suboptimal. To boost efficacy, we targeted the skin layer rich in antigen-presenting cells (APCs) and utilized a jet injector. The jet injection efficiently introduced naked mRNA into skin cells, including APCs in mice. Further analyses indicated that APCs, after taking up antigen mRNA in the skin, migrated to the lymph nodes (LNs) for antigen presentation. Additionally, the jet injection provoked localized lymphocyte infiltration in the skin, serving as a physical adjuvant for vaccination. Without a delivery carrier, our approach confined mRNA distribution to the injection site, preventing systemic mRNA leakage and associated systemic proinflammatory reactions. In mouse vaccination, the naked mRNA jet injection elicited robust antigen-specific antibody production over 6 months, along with germinal center formation in LNs and the induction of both CD4- and CD8-positive T cells. By targeting the SARS-CoV-2 spike protein, this approach provided protection against viral challenge. Furthermore, our approach generated neutralizing antibodies against SARS-CoV-2 in non-human primates at levels comparable to those observed in mice. In conclusion, our approach offers a safe and effective option for mRNA vaccines targeting infectious diseases.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Vacunas de ARNm , Animales , Ratones , SARS-CoV-2/inmunología , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Vacunas de ARNm/inmunología , COVID-19/prevención & control , COVID-19/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Antivirales/inmunología , Femenino , Células Presentadoras de Antígenos/inmunología , ARN Mensajero/genética , ARN Mensajero/inmunología , Linfocitos T CD8-positivos/inmunología , Anticuerpos Neutralizantes/inmunología , Humanos , Vacunación/métodos
2.
Angew Chem Int Ed Engl ; : e202404972, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38651732

RESUMEN

Controlling the end-groups of biocompatible polymers is crucial for enabling polymer-based therapeutics and nanomedicine. Typically, end-group diversification is a challenging and time-consuming endeavor, especially for polymers pre-pared via ionic polymerization mechanisms with limited functional group tolerance. In this study, we present a facile end-group diversification approach for poly(2-oxazoline)s (POx), enabling quick and reliable production of hetero-telechelic polymers to facilitate POxylation. The approach relies on the careful tuning of reaction parameters to establish differential reactivity of a pentafluorobenzyl initiator fragment and the living oxazolinium chain-end, allowing the selective introduction of N-, S-, O-nucleophiles via the termination of the polymerization, and a consecutive nucleophilic para-fluoro substitution. The value of this approach for the accelerated development of nanomedicine is demonstrated through the synthesis of well-defined lipid-polymer conjugates and POx-polypeptide block-copolymers, which are well-suited for drug and gene delivery. Furthermore, we investigated the application of a lipid-POx conjugate for the formulation and delivery of mRNA-loaded lipid nanoparticles for immunization against the SARS-COV-2 virus, underscoring the value of POx as a biocompatible polymer platform.

3.
Cancer Sci ; 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38558246

RESUMEN

Chemoresistance is a major cause of high mortality and poor survival in patients with ovarian cancer (OVCA). Understanding the mechanisms of chemoresistance is urgently required to develop effective therapeutic approaches to OVCA. Here, we show that expression of the long noncoding RNA, taurine upregulated gene 1 (TUG1), is markedly upregulated in samples from OVCA patients who developed resistance to primary platinum-based therapy. Depletion of TUG1 increased sensitivity to cisplatin in the OVCA cell lines, SKOV3 and KURAMOCHI. Combination therapy of cisplatin with antisense oligonucleotides targeting TUG1 coupled with a drug delivery system effectively relieved the tumor burden in xenograft mouse models. Mechanistically, TUG1 acts as a competing endogenous RNA by downregulating miR-4687-3p and miR-6088, both of which target DNA polymerase eta (POLH), an enzyme required for translesion DNA synthesis. Overexpression of POLH reversed the effect of TUG1 depletion on cisplatin-induced cytotoxicity. Our data suggest that TUG1 upregulation allows OVCA to tolerate DNA damage via upregulation of POLH; this provides a strong rationale for targeting TUG1 to overcome cisplatin resistance in OVCA.

4.
Angew Chem Int Ed Engl ; 63(14): e202317817, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38342757

RESUMEN

The differential enzymatic activity in the endo/lysosomes of particular cells could trigger targeted endosomal escape functions, enabling selective intracellular protein delivery. However, this strategy may be jeopardized due to protein degradation during endosomal trafficking. Herein, using custom made fluorescent probes to assess the endosomal activity of cathepsin B (CTSB) and protein degradation, we found that certain cancer cells with hyperacidified endosomes grant a spatiotemporal window where CTSB activity surpass protein digestion. This inspired the engineering of antibody-loaded polymeric nanocarriers having CTSB-activatable endosomal escape ability. The nanocarriers selectively escaped from the endo/lysosomes in the cells with high endosomal CTSB activity and delivered active antibodies to intracellular targets. This study provides a viable strategy for cell-specific protein delivery using stimuli-responsive nanocarriers with controlled endosomal escape.


Asunto(s)
Endosomas , Neoplasias , Endosomas/metabolismo , Anticuerpos/metabolismo , Polímeros/metabolismo , Lisosomas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
5.
Fluids Barriers CNS ; 20(1): 88, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38053174

RESUMEN

Current strategies to identify ligands for brain delivery select candidates based on preferential binding to cell-membrane components (CMC) on brain endothelial cells (EC). However, such strategies generate ligands with inherent brain specificity limitations, as the CMC (e.g., the transferrin receptor TfR1) are also significantly expressed on peripheral EC. Therefore, novel strategies are required to identify molecules allowing increased specificity of therapy brain delivery. Here, we demonstrate that, while individual CMC are shared between brain EC and peripheral EC, their endocytic internalization rate is markedly different. Such differential endocytic rate may be harnessed to identify molecular tags for brain targeting based on their selective retention on the surface of brain EC, thereby generating 'artificial' targets specifically on the brain vasculature. By quantifying the retention of labelled proteins on the cell membrane, we measured the general endocytic rate of primary brain EC to be less than half that of primary peripheral (liver and lung) EC. In addition, through bio-panning of phage-displayed peptide libraries, we unbiasedly probed the endocytic rate of individual CMC of liver, lung and brain endothelial cells. We identified phage-displayed peptides which bind to CMC common to all three endothelia phenotypes, but which are preferentially endocytosed into peripheral EC, resulting in selective retention on the surface of brain EC. Furthermore, we demonstrate that the synthesized free-form peptides are capable of generating artificial cell-surface targets for the intracellular delivery of model proteins into brain EC with increasing specificity over time. The developed identification paradigm, therefore, demonstrates that the lower endocytic rate of individual CMC on brain EC can be harnessed to identify peptides capable of generating 'artificial' targets for the selective delivery of proteins into the brain vasculature. In addition, our approach identifies brain-targeting peptides which would have been overlooked by conventional identification strategies, thereby increasing the repertoire of candidates to achieve specific therapy brain delivery.


Asunto(s)
Encéfalo , Células Endoteliales , Células Endoteliales/metabolismo , Endotelio/metabolismo , Encéfalo/metabolismo , Péptidos/metabolismo
6.
Biomacromolecules ; 24(12): 5915-5925, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-37987713

RESUMEN

Aiming toward the development of tailored carrier materials for the cytostatics panobinostat and imatinib, an amphiphilic block copolymer composed of poly(2-ethyl-2-oxazoline) and a degradable poly(2-(3-phenylpropyl)-2-oxazoline) analogue (dPPhPrOx-b-PEtOx) was synthesized via a postpolymerization synthesis route based on reacylation of oxidized linear poly(ethylene imine). The obtained dPPhPrOx-b-PEtOx was found to readily self-assemble into well-defined micelles with a critical micelle concentration of 1 µg mL-1. The incubation of HUVEC cells with the blank micelles revealed their excellent cytocompatibility (up to 2 mg mL-1), thus confirming the polymers' suitability for potential drug delivery application. Subsequently, the encapsulation of the two cytostatics, panobinostat and imatinib, into the dPPhPrOx-b-PEtOx micelles was successfully demonstrated (Dh ≈ 80 nm, PDI ≈ 0.16), whereby the well-defined nature of the micelle was maintained upon extended incubation at 37 °C (36 h) and storage at 4 °C (1 month). Labeling of the micelles with Alexa Fluor 594 and Alexa Fluor 647, which form a Förster resonance energy transfer (FRET) pair, indicated the stability of loaded micelles upon dilution until the CMC. Finally, the cytotoxicity of the loaded micelles was investigated against three different cell lines: Medulloblastoma cell lines ONS-76 and DAOY as well as the glioblastoma cell line U87MG. While the panobinostat-loaded micelles displayed similar cytotoxicity compared to the pure drug in the cell lines, imatinib-loaded micelles were found to be more potent compared to the pristine drug, as significantly higher cytotoxicity was observed across all three cell lines.


Asunto(s)
Portadores de Fármacos , Micelas , Panobinostat/farmacología , Mesilato de Imatinib/farmacología , Portadores de Fármacos/química , Glicina , Polímeros/química , Polietilenglicoles/química
8.
Nat Commun ; 14(1): 4521, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37607907

RESUMEN

Oncogene-induced DNA replication stress (RS) and consequent pathogenic R-loop formation are known to impede S phase progression. Nonetheless, cancer cells continuously proliferate under such high-stressed conditions through incompletely understood mechanisms. Here, we report taurine upregulated gene 1 (TUG1) long noncoding RNA (lncRNA), which is highly expressed in many types of cancers, as an important regulator of intrinsic R-loop in cancer cells. Under RS conditions, TUG1 is rapidly upregulated via activation of the ATR-CHK1 signaling pathway, interacts with RPA and DHX9, and engages in resolving R-loops at certain loci, particularly at the CA repeat microsatellite loci. Depletion of TUG1 leads to overabundant R-loops and enhanced RS, leading to substantial inhibition of tumor growth. Our data reveal a role of TUG1 as molecule important for resolving R-loop accumulation in cancer cells and suggest targeting TUG1 as a potent therapeutic approach for cancer treatment.


Asunto(s)
Neoplasias , Estructuras R-Loop , Humanos , Replicación del ADN/genética , Proliferación Celular/genética , Neoplasias/genética , Repeticiones de Microsatélite/genética , Taurina
9.
Pharmaceutics ; 15(7)2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37514151

RESUMEN

Large-area craniofacial defects remain a challenge for orthopaedists, hastening the need to develop a facile and safe tissue engineering strategy; osteoconductive material and a combination of optimal growth factors and microenvironment should be considered. Faced with the unmet need, we propose that abundant cytokines and chemokines can be secreted from the bone defect, provoking the infiltration of endogenous stem cells to assist bone regeneration. We can provide a potent mRNA medicine cocktail to promptly initiate the formation of bone templates, osteogenesis, and subsequent bone matrix deposition via endochondral ossification, which may retard rapid fibroblast infiltration and prevent the formation of atrophic non-union. We explored the mutual interaction of BMP2 and TGFß3 mRNA, both potent chondrogenic factors, on inducing endochondral ossification; examined the influence of in vitro the transcribed polyA tail length on mRNA stability; prepared mRNA nanomedicine using a PEGylated polyaspartamide block copolymer loaded in a gelatin sponge and grafted in a critical-sized calvarial defect; and evaluated bone regeneration using histological and µCT examination. The BMP2 and TGFß3 composite mRNA nanomedicine resulted in over 10-fold new bone volume (BV) regeneration in 8 weeks than the BMP2 mRNA nanomedicine administration alone, demonstrating that the TGFß3 mRNA nanomedicine synergistically enhances the bone's formation capability, which is induced by BMP2 mRNA nanomedicine. Our data demonstrated that mRNA-medicine-mediated endochondral ossification provides an alternative cell-free tissue engineering methodology for guiding craniofacial defect healing.

10.
Proc Natl Acad Sci U S A ; 120(29): e2214320120, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37428918

RESUMEN

Integrating antigen-encoding mRNA (Messenger RNA) and immunostimulatory adjuvant into a single formulation is a promising approach to potentiating the efficacy of mRNA vaccines. Here, we developed a scheme based on RNA engineering to integrate adjuvancy directly into antigen-encoding mRNA strands without hampering the ability to express antigen proteins. Short double-stranded RNA (dsRNA) was designed to target retinoic acid-inducible gene-I (RIG-I), an innate immune receptor, for effective cancer vaccination and then tethered onto the mRNA strand via hybridization. Tuning the dsRNA structure and microenvironment by changing its length and sequence enabled the determination of the structure of dsRNA-tethered mRNA efficiently stimulating RIG-I. Eventually, the formulation loaded with dsRNA-tethered mRNA of the optimal structure effectively activated mouse and human dendritic cells and drove them to secrete a broad spectrum of proinflammatory cytokines without increasing the secretion of anti-inflammatory cytokines. Notably, the immunostimulating intensity was tunable by modulating the number of dsRNA along the mRNA strand, which prevents excessive immunostimulation. Versatility in the applicable formulation is a practical advantage of the dsRNA-tethered mRNA. Its formulation with three existing systems, i.e., anionic lipoplex, ionizable lipid-based lipid nanoparticles, and polyplex micelles, induced appreciable cellular immunity in the mice model. Of particular interest, dsRNA-tethered mRNA encoding ovalbumin (OVA) formulated in anionic lipoplex used in clinical trials exerted a significant therapeutic effect in the mouse lymphoma (E.G7-OVA) model. In conclusion, the system developed here provides a simple and robust platform to supply the desired intensity of immunostimulation in various formulations of mRNA cancer vaccines.


Asunto(s)
Neoplasias , ARN Bicatenario , Humanos , Animales , Ratones , ARN Bicatenario/genética , Adyuvantes Inmunológicos/farmacología , Antígenos , Inmunidad Celular , Citocinas/genética , ARN Mensajero/genética , Ratones Endogámicos C57BL , Neoplasias/terapia
11.
J Nanobiotechnology ; 21(1): 36, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36721182

RESUMEN

BACKGROUND: Although a large amount of evidence has revealed that amyloid ß (Aß), especially Aß oligomers, protofibrils, and pyroglutamated Aßs, participate primarily in the pathophysiological processes of Alzheimer's disease, most clinical trials of anti-Aß antibody therapy have never acquired successful efficacy in human clinical trials, partly because peripheral administration of antibody medications was unable to deliver sufficient amounts of the molecules to the brain. Recently, we developed polymeric nanomicelles capable of passing through the blood-brain barrier that function as chaperones to deliver larger amounts of heavy molecules to the brain. Herein, we aimed to evaluate the efficacy of newly developed antibody 6H4 fragments specific to Aß oligomers encapsulated in polymeric nanomicelles on the development of Alzheimer's disease pathology in Alzheimer's disease model mice at the age of emergence of early Alzheimer's disease pathology. RESULTS: During the 10-week administration of 6H4 antibody fragments in polymeric nanomicelles, a significant reduction in the amounts of various toxic Aß species, such as Aß oligomers, toxic Aß conformers, and pyroglutamated Aßs in the brain was observed. In addition, immunohistochemistry indicated inhibition of diameters of Aß plaques, Aß-antibody immunoreactive areas, and also plaque core formation. Behavioral analysis of the mice model revealed that the 6H4 fragments-polymeric nanomicelle group was significantly better at maintaining long-term spatial reference memory in the probe and platform tests of the water maze, thereby indicating inhibition of the pathophysiological process of Alzheimer's disease. CONCLUSIONS: The results indicated that the strategy of reducing toxic Aß species in early dementia owing to Alzheimer's disease by providing sufficient antibodies in the brain may modify Alzheimer's disease progression.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Animales , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo , Barrera Hematoencefálica , Anticuerpos , Placa Amiloide , Polímeros
12.
Adv Sci (Weinh) ; 10(10): e2205139, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36739605

RESUMEN

Treatment of immunologically cold tumors is a major challenge for immune checkpoint inhibitors (ICIs). Interleukin 12 (IL-12) can invigorate ICIs against cold tumors by establishing a robust antitumor immunity. However, its toxicity and systemic induction of counteracting immunosuppressive signals have hindered translation. Here, IL-12 activity is spatiotemporally controlled for safely boosting efficacy without the stimulation of interfering immune responses by generating a nanocytokine that remains inactive at physiological pH, but unleashes its full activity at acidic tumor pH. The IL-12-based nanocytokine (Nano-IL-12) accumulate and release IL-12 in tumor tissues, eliciting localized antitumoral inflammation, while preventing systemic immune response, counteractive immune reactions, and adverse toxicities even after repeated intravenous administration. The Nano-IL-12-mediated spatiotemporal control of inflammation prompt superior anticancer efficacy, and synergize with ICIs to profoundly inflame the tumor microenvironment and completely eradicate ICI-resistant primary and metastatic tumors. The strategy could be a promising approach toward safer and more effective immunotherapies.


Asunto(s)
Interleucina-12 , Neoplasias , Humanos , Neoplasias/terapia , Inflamación/patología , Inmunoterapia , Microambiente Tumoral
13.
Biomater Sci ; 11(7): 2336-2347, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36804651

RESUMEN

Targeting brain lipid metabolism is a promising strategy to regulate the energy balance and fight metabolic diseases such as obesity. The development of stable platforms for selective delivery of drugs, particularly to the hypothalamus, is a challenge but a possible solution for these metabolic diseases. Attenuating fatty acid oxidation in the hypothalamus via CPT1A inhibition leads to satiety, but this target is difficult to reach in vivo with the current drugs. We propose using an advanced crosslinked polymeric micelle-type nanomedicine that can stably load the CPT1A inhibitor C75-CoA for in vivo control of the energy balance. Central administration of the nanomedicine induced a rapid attenuation of food intake and body weight in mice via regulation of appetite-related neuropeptides and neuronal activation of specific hypothalamic regions driving changes in the liver and adipose tissue. This nanomedicine targeting brain lipid metabolism was successful in the modulation of food intake and peripheral metabolism in mice.


Asunto(s)
Metabolismo de los Lípidos , Nanomedicina , Ratones , Animales , Metabolismo Energético , Obesidad/metabolismo , Hipotálamo/metabolismo
15.
Nat Commun ; 13(1): 7165, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36418896

RESUMEN

Nano-immunotherapy improves breast cancer outcomes but not all patients respond and none are cured. To improve efficacy, research focuses on drugs that reprogram cancer-associated fibroblasts (CAFs) to improve therapeutic delivery and immunostimulation. These drugs, however, have a narrow therapeutic window and cause adverse effects. Developing strategies that increase CAF-reprogramming while limiting adverse effects is urgent. Here, taking advantage of the CAF-reprogramming capabilities of tranilast, we developed tranilast-loaded micelles. Strikingly, a 100-fold reduced dose of tranilast-micelles induces superior reprogramming compared to free drug owing to enhanced intratumoral accumulation and cancer-associated fibroblast uptake. Combination of tranilast-micelles and epirubicin-micelles or Doxil with immunotherapy increases T-cell infiltration, resulting in cures and immunological memory in mice bearing immunotherapy-resistant breast cancer. Furthermore, shear wave elastography (SWE) is able to monitor reduced tumor stiffness caused by tranilast-micelles and predict response to nano-immunotherapy. Micellar encapsulation is a promising strategy for TME-reprogramming and SWE is a potential biomarker of response.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Neoplasias , Ratones , Animales , Micelas , Microambiente Tumoral , Inmunoterapia , ortoaminobenzoatos/farmacología , ortoaminobenzoatos/uso terapéutico , Factores Inmunológicos , Polímeros
16.
ACS Nano ; 16(8): 12290-12304, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35942986

RESUMEN

Assessment of drug activation and subsequent interaction with targets in living tissues could guide nanomedicine design, but technologies enabling insight into how a drug reaches and binds its target are limited. We show that a Hoechst-based reporter system can monitor drug release and engagement from a nanoparticle delivery system in vitro and in vivo, elucidating differences in target-bound drug distribution related to drug-linker and nanoparticle properties. Drug engagement is defined as chemical detachment of drug or reporter from a nanoparticle and subsequent binding to a subcellular target, which in the case of Hoechst results in a fluorescence signal. Hoechst-based nanoreporters for drug activation contain prodrug elements such as dipeptide linkers, conjugation handles, and nanoparticle modifications such as targeting ligands to determine how nanomedicine design affects distribution of drug engaged with a subcellular target, which is tracked via cellular nuclear fluorescence in situ. Furthermore, the nanoplatform is amenable toward common maleimide-based linkers found in many prodrug-based delivery systems including polymer-, peptide-, and antibody-drug conjugates. Findings from the Hoechst reporter system were applied to develop highly potent, targeted, anticancer micelle nanoparticles delivering a monomethyl auristatin E (MMAE) prodrug comprising the same linkers employed in Hoechst studies. MMAE nanomedicine with the optimal drug-linker resulted in effective tumor growth inhibition in mice without associated acute toxicity, whereas the nonoptimal linker that showed broader drug activation in Hoechst reporter studies resulted in severe toxicity. Our results demonstrate the potential to synergize direct visualization of drug engagement with nanomedicine drug-linker design to optimize safety and efficacy.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Nanopartículas , Profármacos , Ratones , Animales , Profármacos/química , Ensayos Antitumor por Modelo de Xenoinjerto , Inmunoconjugados/química , Micelas , Nanopartículas/uso terapéutico , Nanopartículas/química , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Sistemas de Liberación de Medicamentos
17.
FASEB J ; 36(9): e22486, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35929425

RESUMEN

Neointimal hyperplasia (NIH) after revascularization is a key unsolved clinical problem. Various studies have shown that attenuation of the acute inflammatory response on the vascular wall can prevent NIH. MicroRNA146a-5p (miR146a-5p) has been reported to show anti-inflammatory effects by inhibiting the NF-κB pathway, a well-known key player of inflammation of the vascular wall. Here, a nanomedicine, which can reach the vascular injury site, based on polymeric micelles was applied to deliver miR146a-5p in a rat carotid artery balloon injury model. In vitro studies using inflammation-induced vascular smooth muscle cell (VSMC) was performed. Results showed anti-inflammatory response as an inhibitor of the NF-κB pathway and VSMC migration, suppression of reactive oxygen species production, and proinflammatory cytokine gene expression in VSMCs. A single systemic administration of miR146a-5p attenuated NIH and vessel remodeling in a carotid artery balloon injury model in both male and female rats in vivo. MiR146a-5p reduced proinflammatory cytokine gene expression in injured arteries and monocyte/macrophage infiltration into the vascular wall. Therefore, miR146a-5p delivery to the injury site demonstrated therapeutic potential against NIH after revascularization.


Asunto(s)
Traumatismos de las Arterias Carótidas , MicroARNs , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Arterias , Traumatismos de las Arterias Carótidas/metabolismo , Proliferación Celular , Citocinas/metabolismo , Femenino , Hiperplasia/metabolismo , Inflamación/metabolismo , Masculino , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , FN-kappa B/metabolismo , Nanomedicina , Neointima/tratamiento farmacológico , Neointima/metabolismo , Neointima/prevención & control , Ratas
18.
Biomaterials ; 288: 121748, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36038419

RESUMEN

Intracellular protein delivery is a powerful strategy for developing innovative therapeutics. Nanocarriers present great potential to deliver proteins inside cells by promoting cellular uptake and overcoming entrapment and degradation in acidic endo/lysosomal compartments. Thus, because cytosolic access is essential for eliciting the function of proteins, significant efforts have been dedicated to engineering nanocarriers with maximal endosomal escape regardless of the cell type. On the other hand, controlling the ability of nanocarriers to escape from the endo/lysosomal compartments of particular cells may offer the opportunity for enhancing delivery precision. To test this hypothesis, we developed pH-sensitive polymeric nanocarriers with adjustable endosomal escape potency for selectively reaching the cytosol of defined cancer cells with dysregulated endo/lysosomal acidification. By loading antibodies against nuclear pore complex in the nanocarriers, we demonstrated the selective delivery into the cytosol and subsequent nucleus targeting of cancer cells rather than non-cancerous cells both in vitro and in vivo. Systemically injected nanocarriers loading anti-c-MYC antibodies suppressed c-MYC in solid tumors and inhibit tumor growth without side effects, confirming the therapeutic potential of our approach. These results indicated that regulating the ability of nanocarriers to escape from endo/lysosomal compartments in particular cells is a practical approach for gaining delivery specificity.


Asunto(s)
Nanopartículas , Neoplasias , Citosol/metabolismo , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Endosomas/metabolismo , Humanos , Lisosomas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Polímeros/metabolismo
19.
J Control Release ; 345: 709-720, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35367476

RESUMEN

Since its license in 1978, cisplatin has proved to be one of the most successful chemotherapeutic agents in the world. However, two acute challenges facing cisplatin, resistance and toxicity, have resulted in a bottleneck of clinical application. Targeted nanomedicine shows great promise in delivering cisplatin for maximizing efficacy while minimizing off-target toxicity. This article surveyed the recent progress and challenges of targeted nanomedicine in managing resistance and toxicity of cisplatin in both fundamental and clinical aspects. Particularly, we focused on three major mechanisms counteracting cisplatin sensitivity (decreased intracellular accumulation, increased cisplatin deactivation, and enhanced DNA repair/translesion synthesis) and correspondingly highlighted a few representative approaches to increase cisplatin sensitivity through improving the intracellular concentration of cisplatin and implementing combination therapy. Moreover, the requirements for future advancements in cisplatin delivery systems are rendered with emphasis on (i) understanding of nano-bio interaction and post-accumulation biological effects instead of overwhelmingly improving tumor accumulation, (ii) development of stimuli-responsive and/or actively-targeted nanomedicines, (iii) optimization of combination therapy, (iv) novel combinations targeting tumor microenvironment and immunotherapy. We postulate that cisplatin-based nanomedicines will continuously advance and potentially revolutionize oncological treatment.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Inmunoterapia/métodos , Nanomedicina/métodos , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...