Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 272
Filtrar
2.
J Control Release ; 372: 674-681, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38909700

RESUMO

One of the primary obstacles in treating central nervous system (CNS) disorders lies in the limited ability of disease-modifying drugs to cross the blood-brain barrier (BBB). Our previously described Minimally Invasive Nasal Depot (MIND) technique has proven successful in delivering various drugs to the brain in rat models via a trans-olfactory mucosal approach. In this study, we introduce a novel Minimally Invasive Nasal Infusion (MINI) delivery approach for administering ovalbumin, a model protein, utilizing a programmable infusion pump (iPRECIO SMP-310R) in a mouse model. This research highlights the significant role of olfactory mucosa in nose-to-brain delivery, with an efficacy of nearly 45% compared to intracerebroventricular (ICV) administration. This demonstrates its potential as an alternative procedure for treating CNS diseases, offering a greater safety profile relative to the highly invasive clinical routes traditionally adopted for CNS drug delivery.


Assuntos
Administração Intranasal , Ovalbumina , Animais , Ovalbumina/administração & dosagem , Sistemas de Liberação de Medicamentos , Masculino , Camundongos , Encéfalo/metabolismo , Bombas de Infusão , Barreira Hematoencefálica/metabolismo , Camundongos Endogâmicos C57BL
3.
J Control Release ; 372: 661-673, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38936742

RESUMO

Heterobifunctional small molecule degraders are a subset of targeted protein degraders (TPDs), consisting of two ligands joined by a linker to induce proteasomal degradation of a target protein. As compared to traditional small molecules these compounds generally demonstrate inflated physicochemical properties, which may require innovative formulation strategies to enable their delivery and exert pharmacodynamic effect. The blood brain barrier (BBB) serves an essential function in human physiology, but its presence requires advanced approaches for treating central nervous system (CNS) diseases. By integrating emerging modalities like TPDs with conventional concepts of drug delivery, novel strategies to overcome the BBB can be developed. Amongst the available routes, lipid and polymer-based long-acting delivery seems to be the most amenable to TPDs, due to their ability to encapsulate lipophilic cargo and potential to be functionalized for targeted delivery. Another key consideration will be understanding E3 ligase expression in the different regions of the brain. Discovery of new brain or CNS disease specific E3 ligases could help overcome some of the barriers currently associated with CNS delivery of TPDs. This review discusses the current strategies that exist to overcome and improve therapeutic delivery of TPDs to the CNS.


Assuntos
Sistemas de Liberação de Medicamentos , Humanos , Animais , Barreira Hematoencefálica/metabolismo , Proteólise/efeitos dos fármacos , Doenças do Sistema Nervoso Central/tratamento farmacológico , Doenças do Sistema Nervoso Central/metabolismo
4.
Theranostics ; 14(6): 2526-2543, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646640

RESUMO

Rationale: A mature tissue resident macrophage (TRM) population residing in the peritoneal cavity has been known for its unique ability to migrate to peritoneally located injured tissues and impart wound healing properties. Here, we sought to expand on this unique ability of large peritoneal macrophages (LPMs) by investigating whether these GATA6+ LPMs could also intravasate into systemic circulation and migrate to extra-peritoneally located lungs upon ablating lung-resident alveolar macrophages (AMs) by intranasally administered clodronate liposomes in mice. Methods: C12-200 cationic lipidoid-based nanoparticles were employed to selectively deliver a small interfering RNA (siRNA)-targeting CD-45 labeled with a cyanine 5.5 (Cy5.5) dye to LPMs in vivo via intraperitoneal injection. We utilized a non-invasive optical technique called Diffuse In Vivo Flow Cytometry (DiFC) to then systemically track these LPMs in real time and paired it with more conventional techniques like flow cytometry and immunocytochemistry to initially confirm uptake of C12-200 encapsulated siRNA-Cy5.5 (siRNA-Cy5.5 (C12-200)) into LPMs, and further track them from the peritoneal cavity to the lungs in a mouse model of AM depletion incited by intranasally administered clodronate liposomes. Also, we stained for LPM-specific marker zinc-finger transcription factor GATA6 in harvested cells from biofluids like broncho-alveolar lavage as well as whole blood to probe for Cy5.5-labeled LPMs in the lungs as well as in systemic circulation. Results: siRNA-Cy5.5 (C12-200) was robustly taken up by LPMs. Upon depletion of lung-resident AMs, these siRNA-Cy5.5 (C12-200) labeled LPMs rapidly migrated to the lungs via systemic circulation within 12-24 h. DiFC results showed that these LPMs intravasated from the peritoneal cavity and utilized a systemic route of migration. Moreover, immunocytochemical staining of zinc-finger transcription factor GATA6 further confirmed results from DiFC and flow cytometry, confirming the presence of siRNA-Cy5.5 (C12-200)-labeled LPMs in the peritoneum, whole blood and BALF only upon clodronate-administration. Conclusion: Our results indicate for the very first time that selective tropism, migration, and infiltration of LPMs into extra-peritoneally located lungs was dependent on clodronate-mediated AM depletion. These results further open the possibility of therapeutically utilizing LPMs as delivery vehicles to carry nanoparticle-encapsulated oligonucleotide modalities to potentially address inflammatory diseases, infectious diseases and even cancer.


Assuntos
Ácido Clodrônico , Pulmão , Macrófagos Peritoneais , Nanopartículas , Animais , Ácido Clodrônico/farmacologia , Ácido Clodrônico/administração & dosagem , Nanopartículas/química , Nanopartículas/administração & dosagem , Camundongos , Pulmão/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , RNA Interferente Pequeno/administração & dosagem , Fator de Transcrição GATA6/metabolismo , Lipossomos , Camundongos Endogâmicos C57BL , Carbocianinas/química , Movimento Celular/efeitos dos fármacos , Citometria de Fluxo
5.
Adv Drug Deliv Rev ; 208: 115283, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38494152

RESUMO

Neurological disorders are a diverse group of conditions that pose an increasing health burden worldwide. There is a general lack of effective therapies due to multiple reasons, of which a key obstacle is the presence of the blood-brain barrier, which limits drug delivery to the central nervous system, and generally restricts the pool of candidate drugs to small, lipophilic molecules. However, in many cases, these are unable to target key pathways in the pathogenesis of neurological disorders. As a group, RNA therapies have shown tremendous promise in treating various conditions because they offer unique opportunities for specific targeting by leveraging Watson-Crick base pairing systems, opening up possibilities to modulate pathological mechanisms that previously could not be addressed by small molecules or antibody-protein interactions. This potential paradigm shift in disease management has been enabled by recent advances in synthesizing, purifying, and delivering RNA. This review explores the use of RNA-based therapies specifically for central nervous system disorders, where we highlight the inherent limitations of RNA therapy and present strategies to augment the effectiveness of RNA therapeutics, including physical, chemical, and biological methods. We then describe translational challenges to the widespread use of RNA therapies and close with a consideration of future prospects in this field.


Assuntos
Doenças do Sistema Nervoso Central , Nanopartículas , Humanos , RNA/metabolismo , Doenças do Sistema Nervoso Central/tratamento farmacológico , Barreira Hematoencefálica/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética/métodos
6.
Exp Neurol ; 374: 114696, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38244886

RESUMO

Clinical studies have consistently shown that neurodegenerative diseases (NDs) such as Parkinson's disease, Alzheimer's disease, Amyotrophic Lateral Sclerosis, and Huntington's disease show absent or low levels of brain-derived neurotrophic factor (BDNF). Despite this relationship between BDNF and ND, only a few ND animal models have been able to recapitulate the low BDNF state, thereby hindering research into the therapeutic targeting of this important neurotrophic factor. In order to address this unmet need, we sought to develop a reproducible model of BDNF reduction by inducing traumatic brain injury (TBI) using a closed head momentum exchange injury model in mature 9-month-old male and female rats. Head impacts were repetitive and varied in intensity from mild to severe. BDNF levels, as assessed by ELISA, were significantly reduced in the hippocampus of both males and females as well as in the substantia nigra of males 12 days after mild TBI. However, we observed significant sexual dimorphism in multiple sequelae, including magnetic resonance imaging-determined vasogenic edema, astrogliosis (GFAP-activation), and microgliosis (Iba1 activation). This study provides an opportunity to investigate the mechanism of BDNF reduction in rodent models and provides a reliable paradigm to test BDNF-targeted therapeutics for the treatment of ND.


Assuntos
Concussão Encefálica , Lesões Encefálicas Traumáticas , Animais , Feminino , Masculino , Ratos , Concussão Encefálica/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/complicações , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/metabolismo , Substância Negra/metabolismo
7.
Nat Rev Drug Discov ; 23(4): 281-300, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38263456

RESUMO

mRNA formulated with lipid nanoparticles is a transformative technology that has enabled the rapid development and administration of billions of coronavirus disease 2019 (COVID-19) vaccine doses worldwide. However, avoiding unacceptable toxicity with mRNA drugs and vaccines presents challenges. Lipid nanoparticle structural components, production methods, route of administration and proteins produced from complexed mRNAs all present toxicity concerns. Here, we discuss these concerns, specifically how cell tropism and tissue distribution of mRNA and lipid nanoparticles can lead to toxicity, and their possible reactogenicity. We focus on adverse events from mRNA applications for protein replacement and gene editing therapies as well as vaccines, tracing common biochemical and cellular pathways. The potential and limitations of existing models and tools used to screen for on-target efficacy and de-risk off-target toxicity, including in vivo and next-generation in vitro models, are also discussed.


Assuntos
Nanopartículas , Vacinas , Humanos , Vacinas/efeitos adversos , Vacinas contra COVID-19/efeitos adversos , Edição de Genes , Terapia Genética , RNA Mensageiro/genética
8.
Nanomedicine (Lond) ; 18(20): 1399-1415, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37800470

RESUMO

Chronic rhinosinusitis (CRS) is a chronic inflammatory disease of the paranasal sinuses which represents a significant health burden due to its widespread prevalence and impact on patients' quality of life. As the molecular pathways driving and sustaining inflammation in CRS become better elucidated, the diversity of treatment options is likely to widen significantly. Nanotechnology offers several tools to enhance the effectiveness of topical therapies, which has been limited by factors such as poor drug retention, mucosal permeation and adhesion, removal by epithelial efflux pumps and the inability to effectively penetrate biofilms. In this review, we highlight the successful application of nanomedicine in the field of CRS therapeutics, discuss current limitations and propose opportunities for future work.


Chronic sinusitis is a common inflammatory condition of the sinuses, which affects patients' quality of life and consumes significant healthcare resources. It is primarily treated with corticosteroids, a type of medication that reduces inflammation, as a nasal spray or taken orally. Nasal sprays are preferred, to minimize side effects elsewhere in the body. Recently, another class of drugs ­ 'biologic agents' ­ has been approved for a subtype of chronic sinusitis that causes polyps (grape-like swellings of the sinus lining). However, a lasting cure is elusive, because inflammation frequently returns once these medications are stopped. As our understanding of what causes chronic sinusitis improves, researchers are seeking therapies that more accurately target the cause of inflammation, rather than broadly suppressing all types of inflammation using corticosteroids. The use of nanotechnology allows the design of drugs to overcome various challenges in treating chronic sinusitis, potentially enabling more accurate delivery of drugs into the sinuses, improving drugs' ability to remain on the sinus lining and penetrate it, reducing the amount of drug lost due to the action of outflow pumps and overcoming additional defenses built up by bacteria when they form thick films. Here, we describe how nanomedicine has been used to develop drugs for chronic sinusitis, discuss current limitations and propose opportunities for future work.


Assuntos
Seios Paranasais , Rinite , Sinusite , Humanos , Qualidade de Vida , Rinite/tratamento farmacológico , Rinite/metabolismo , Sinusite/tratamento farmacológico , Sinusite/metabolismo , Seios Paranasais/metabolismo , Doença Crônica , Nanotecnologia
9.
J Control Release ; 363: 435-451, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37717658

RESUMO

It is time for the story of mitochondria and intracellular communication in multidrug resistant cancer to be rewritten. Herein we characterize the extent and cellular advantages of mitochondrial network fusion in multidrug resistant (MDR) breast cancer and have designed a novel nanomedicine that disrupts mitochondrial network fusion and systematically manipulates organelle fusion and function. Combination Organelle Mitochondrial Endoplasmic reticulum Therapy (COMET) is an innovative translational nanomedicine for treating MDR triple negative breast cancer (TNBC) that has superior safety and equivalent efficacy to the current standard of care (paclitaxel). Our study has demonstrated that the increased mitochondrial networks in MDR TNBC contribute to apoptotic resistance and network fusion is mediated by mitofusin2 (MFN2) on the outer mitochondrial membrane. COMET consists of three components; Mitochondrial Network Disrupting (MiND) nanoparticles (NPs) that are loaded with an anti-MFN2 peptide, tunicamycin, and Bam7. The therapeutic rationale of COMET is to reduce the apoptotic threshold in MDR cells with MiND NPs, followed by inducing the endoplasmic reticulum mediated unfolded protein response (UPR) by stressing MDR cells with tunicamycin, and finally, directly inducing mitochondrial apoptosis with Bam7 which is a specific bcl-2 Bax activator. MiND NPs are PEGylated liposomes with the 21 amino acid (2577.98 MW) anti-MFN2 peptide compartmentalized in the aqueous core. Hypoxia (0.5% oxygen) was used to create MDR derivatives of MDA-MB-231 cells and BT-549 cells. Mitochondrial networks were quantified using 3D analysis of 60× live cell images acquired with a Keyence BZ-X710 microscope and MiND NPs effectively fragmented mitochondrial networks in drug sensitive and MDR TNBC cells. The IC50 values, combination index, and dose reduction index derived from dose response studies demonstrate that MiND NPs decrease the apoptotic threshold of both drug sensitive and MDR TNBC cells and COMET is a synergistic drug combination. Complex V (ATP synthase) extracted from bovine cardiac mitochondria was used to assess the effect of MiND NPs on OXPHOS; both MiND NPs and anti-MFN2 peptide solution significantly decrease the activity of mitochondrial complex V and decrease the capacity of OXPHOS. A BacMam viral vector based fluorescent biosensor was used to quantify the unfolded protein response (UPR) at the level of the endoplasmic reticulum and tunicamycin specifically induces the UPR in drug sensitive and MDR TNBC cells. A caspase 3 colorimetric assay demonstrated that the synergistic triple drug combination of COMET increases the ability of Bam7 to specifically induce apoptosis. Dose limiting toxicity and off target effects are a significant challenge for current chemotherapy regimens including paclitaxel. COMET has significantly lower cytotoxicity than paclitaxel in human embryonic kidney epithelial cells and has the potential to fulfill the clinical need for safer cancer therapeutics. COMET is a promising early stage translational nanomedicine for treating MDR TNBC. Manipulating intracellular communication and organelle fusion is a novel approach to treating MDR cancer. The data from this study has rewritten the story of mitochondria, organelle fusion, and intracellular communication and by targeting this intersection, COMET is an exciting new chapter in cancer therapeutics that could transform the clinical outcome of MDR TNBC.


Assuntos
Resistência a Múltiplos Medicamentos , Neoplasias de Mama Triplo Negativas , Animais , Bovinos , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Tunicamicina/metabolismo , Tunicamicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Paclitaxel , Mitocôndrias , Apoptose , Retículo Endoplasmático/metabolismo , Peptídeos/farmacologia , Combinação de Medicamentos , Linhagem Celular Tumoral
10.
Adv Drug Deliv Rev ; 197: 114853, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37149040

RESUMO

Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.


Assuntos
Glioma , Humanos , Glioma/tratamento farmacológico , Sistemas de Liberação de Medicamentos
11.
Expert Opin Drug Deliv ; 20(4): 489-506, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36890642

RESUMO

INTRODUCTION: In November 2019, the idea of a zoonotic virus crossing over to human transmission in a seafood market in Wuhan, China, and then soaring across the globe to claim over 6.3 million lives and persisting to date, seemed more like wild science fiction than a future reality. As the SARS-CoV-2 pandemic continues, it is important to hallmark the imprints the pandemic has made on science. AREAS COVERED: This review covers the biology of SARS-CoV-2, vaccine formulations and trials, the concept of 'herd resistance,' and the vaccination divide. EXPERT OPINION: The SARS-CoV-2 pandemic has changed the landscape of medicine. The rapid approval of SARS-CoV-2 vaccines has changed the culture of drug development and clinical approvals. This change is already leading to more accelerated trials. The RNA vaccines have opened the market for nucleic acid therapies and the applications are limitless - from cancer to influenza. A phenomenon that has occurred is that the low efficacy of current vaccines and the rapid mutation rate of the virus is preventing herd immunity from being attained. Instead, herd resistance is being acquired. Even with future, more effective vaccines, anti-vaccination attitudes will continue to challenge the quest for SARS-CoV-2 herd immunity.


Assuntos
COVID-19 , Ácidos Nucleicos , Humanos , Pandemias/prevenção & controle , SARS-CoV-2 , Vacinas contra COVID-19 , Imunidade Coletiva , Nanomedicina , COVID-19/prevenção & controle
12.
Mol Pharm ; 20(3): 1729-1736, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36744718

RESUMO

Verapamil is a calcium channel blocker that holds promise for the therapy of chronic rhinosinusitis (CRS) with and without nasal polyps. The verapamil-induced side effects limit its tolerated dose via the oral route, underscoring the usefulness of localized intranasal administration. However, the challenge to intranasal administration is mucociliary clearance, which diminishes localized dose availability. To overcome this challenge, verapamil was loaded into a mucoadhesive cationic poly(ethylene glycol)-modified (PEGylated) liposomal carrier. Organotypic nasal explants were exposed to verapamil liposomes under flow conditions to mimic mucociliary clearance. The liposomes resulted in significantly higher tissue residence compared with the free verapamil control. These findings were further confirmed in vivo in C57BL/6 mice following intranasal administration. Liposomes significantly increased the accumulation of verapamil in nasal tissues compared with the control group. The developed tissue-retentive verapamil liposomal formulation is considered a promising intranasal delivery system for CRS therapy.


Assuntos
Lipossomos , Sinusite , Animais , Camundongos , Lipossomos/uso terapêutico , Verapamil , Polietilenoglicóis/uso terapêutico , Camundongos Endogâmicos C57BL , Administração Intranasal , Sinusite/tratamento farmacológico , Administração Tópica
13.
Pharmaceutics ; 15(1)2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36678865

RESUMO

The development of new vaccine adjuvants represents a key approach to improvingi the immune responses to recombinant vaccine antigens. Emulsion adjuvants, such as AS03 and MF59, in combination with influenza vaccines, have allowed antigen dose sparing, greater breadth of responses and fewer immunizations. It has been demonstrated previously that emulsion adjuvants can be prepared using a simple, low-shear process of self-emulsification (SE). The role of alpha tocopherol as an immune potentiator in emulsion adjuvants is clear from the success of AS03 in pandemic responses, both to influenza and COVID-19. Although it was a significant formulation challenge to include alpha tocopherol in an emulsion prepared by a low-shear process, the resultant self-emulsifying adjuvant system (SE-AS) showed a comparable effect to the established AS03 when used with a quadrivalent influenza vaccine (QIV). In this paper, we first optimized the SE-AS with alpha tocopherol to create SE-AS44, which allowed the emulsion to be sterile-filtered. Then, we compared the in vitro cell activation cytokine profile of SE-AS44 with the self-emulsifying adjuvant 160 (SEA160), a squalene-only adjuvant. In addition, we evaluated SE-AS44 and SEA160 competitively, in combination with a recombinant cytomegalovirus (CMV) pentamer antigen mouse.

14.
J Allergy Clin Immunol ; 151(2): 509-525.e8, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36494212

RESUMO

BACKGROUND: The human upper respiratory tract is the first site of contact for inhaled respiratory viruses and elaborates an array of innate immune responses. Seasonal variation in respiratory viral infections and the importance of ambient temperature in modulating immune responses to infections have been well recognized; however, the underlying biological mechanisms remain understudied. OBJECTIVE: We investigated the role of nasal epithelium-derived extracellular vesicles (EVs) in innate Toll-like receptor 3 (TLR3)-dependent antiviral immunity. METHODS: We evaluated the secretion and composition of nasal epithelial EVs after TLR3 stimulation in human autologous cells and fresh human nasal mucosal surgical specimens. We also explored the antiviral activity and mechanisms of TLR3-stimulated EVs against respiratory viruses as well as the effect of cool ambient temperature on TLR3-dependent antiviral immunity. RESULTS: We found that polyinosinic:polycytidylic acid, aka poly(I:C), exposure induced a swarm-like increase in the secretion of nasal epithelial EVs via the TLR3 signaling. EVs participated in TLR3-dependent antiviral immunity, protecting the host from viral infections through both EV-mediated functional delivery of miR-17 and direct virion neutralization after binding to virus ligands via surface receptors, including LDLR and ICAM-1. These potent antiviral immune defense functions mediated by TLR3-stimulated EVs were impaired by cold exposure via a decrease in total EV secretion as well as diminished microRNA packaging and antiviral binding affinity of individual EV. CONCLUSION: TLR3-dependent nasal epithelial EVs exhibit multiple innate antiviral mechanisms to suppress respiratory viral infections. Furthermore, our study provides a direct quantitative mechanistic explanation for seasonal variation in upper respiratory tract infection prevalence.


Assuntos
Vesículas Extracelulares , Viroses , Humanos , Receptor 3 Toll-Like , Imunidade Inata , Antivirais/farmacologia , Poli I-C
15.
Pharm Res ; 40(1): 77-105, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36380168

RESUMO

Nucleic acid-based therapeutic molecules including small interfering RNA (siRNA), microRNA(miRNA), antisense oligonucleotides (ASOs), messenger RNA (mRNA), and DNA-based gene therapy have tremendous potential for treating diseases in the central nervous system (CNS). However, achieving clinically meaningful delivery to the brain and particularly to target cells and sub-cellular compartments is typically very challenging. Mediating cell-specific delivery in the CNS would be a crucial advance that mitigates off-target effects and toxicities. In this review, we describe these challenges and provide contemporary evidence of advances in cellular and sub-cellular delivery using a variety of delivery mechanisms and alternative routes of administration, including the nose-to-brain approach. Strategies to achieve subcellular localization, endosomal escape, cytosolic bioavailability, and nuclear transfer are also discussed. Ultimately, there are still many challenges to translating these experimental strategies into effective and clinically viable approaches for treating patients.


Assuntos
Sistemas de Liberação de Medicamentos , MicroRNAs , Ácidos Nucleicos , RNA Interferente Pequeno , Humanos , Barreira Hematoencefálica , Encéfalo , MicroRNAs/uso terapêutico , Ácidos Nucleicos/uso terapêutico , Oligonucleotídeos Antissenso/uso terapêutico , RNA Interferente Pequeno/uso terapêutico
16.
Bioact Mater ; 22: 551-566, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36382022

RESUMO

Cancer therapy is undergoing a paradigm shift toward immunotherapy focusing on various approaches to activate the host immune system. As research to identify appropriate immune cells and activate anti-tumor immunity continues to expand, scientists are looking at microbial sources given their inherent ability to elicit an immune response. Bacterial extracellular vesicles (BEVs) are actively studied to control systemic humoral and cellular immune responses instead of using whole microorganisms or other types of extracellular vesicles (EVs). BEVs also provide the opportunity as versatile drug delivery carriers. Unlike mammalian EVs, BEVs have already made it to the clinic with the meningococcal vaccine (Bexsero®). However, there are still many unanswered questions in the use of BEVs, especially for chronic systemically administered immunotherapies. In this review, we address the opportunities and challenges in the use of BEVs for cancer immunotherapy and provide an outlook towards development of BEV products that can ultimately translate to the clinic.

17.
Nucleic Acid Ther ; 33(1): 35-44, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36318487

RESUMO

Despite wide recognition as a disease of pandemic proportions, effective treatments for nonalcoholic fatty liver disease (NAFLD) remain elusive. Most of the current clinical programs aim to reduce hepatic fat accumulation and, thus, prevent downstream inflammation and fibrosis. To date, this therapeutic approach has helped identify a potential disconnect between steatosis reduction and disease resolution. Mounting preclinical evidence indicates liver inflammation may play a major role in steatosis development and fibrosis but has not garnered the same clinical representation. This may be owing to deficiencies in standard therapeutic modalities that limit their application in NAFLD. RNA interference (RNAi) is an attractive approach to targeting liver inflammation owing to its clinical safety profile, target specificity, and limited biodistribution. In this study, we characterize a simple cholesterol-short-interfering RNA (siRNA) conjugate system targeting Tnf mRNA in liver macrophages for the treatment of NAFLD. First, we observed delivery and anti-inflammatory activity in an acute liver inflammation model. In a follow-up murine NAFLD model, we observed total prevention of nearly all hallmarks of this disease: steatosis, inflammation, and fibrosis. This simple conjugate siRNA delivery system may be the first to show RNAi activity in liver macrophages and provide evidence for a novel therapeutic approach to inflammation in NAFLD.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/terapia , Hepatopatia Gordurosa não Alcoólica/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Distribuição Tecidual , Fígado/metabolismo , Células de Kupffer , Inflamação/genética , Inflamação/terapia , Colesterol , RNA de Cadeia Dupla/metabolismo , Fibrose
18.
Biomater Sci ; 11(1): 181-194, 2022 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-36408841

RESUMO

Epidermal growth factor (EGF) is required for various regulations of skin tissue including wound healing; however, it has limited stability due to the physicochemical conditions of the wound milieu. The lack of functional EGF within the wound can cause permanent tissue defects and therefore, current wound patch designs involve EGF-releasing components. Consequently, the focus of such systems is to improve the wound healing mechanism, with minimal attention on melanogenesis of the scar tissue. The present study investigates in vitro/in vivo wound healing and melanogenesis potential of the EGF-doped films comprised of arrays of chitosan:gelatin nanopillars (nano C:G films) prepared by using nanoporous anodic alumina molds. The potential of EGF-doped films in wound healing was examined with individual and coculture systems of fibroblasts and melanocytes to mimic the wound conditions. The outcomes demonstrated that compared to the control groups, the combination of EGF doping and nanotopography consistently provided the highest levels of melanogenic activity-related genes, melanin contents as well as EGFR expressions for both melanocyte-only and coculture setups. Proteomic, genomic and histological analysis of the excisional wound model further demonstrated that if EGF was present within the nanostructured films, the performance of these substrates in terms of wound closure, collagen thickness as well as melanin deposition was considerably improved. Furthermore, when compared with the control saline treatment and healthy mice groups, significant differences for such parameters were obtained for the nano C:G films, irrespective of their EGF contents. Overall, the results indicate that EGF-doped nano C:G films are good candidates as wound patches that not only provide desirable healing characteristics but also cause improved melanogenic outputs.


Assuntos
Quitosana , Fator de Crescimento Epidérmico , Camundongos , Animais , Fator de Crescimento Epidérmico/metabolismo , Gelatina , Quitosana/química , Melaninas , Proteômica , Cicatrização
19.
Mol Pharm ; 19(11): 4067-4079, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36226722

RESUMO

Extracellular vesicle (EV)-mediated microRNA transfer and propagation from the donor cell to the recipient cell in the tumor microenvironment have significant implications, including the development of multidrug resistance (MDR). Although miRNA-encapsulated EV have been shown to have functional effects on recipient cells, the quantitative aspects of transfer kinetics and functional effects remain poorly understood. Intracellular events such as degradation of miRNA, loading of miRNA into EVs, cellular release of EVs, and their uptake by recipient cells govern the transfer and functional effect of encapsulated miRNA. Based on these rate-limiting steps, we developed a mathematical model using ordinary differential equations (model 1). We performed coculture experiments using ID8-VEGF ovarian cancer cells to demonstrate EV-mediated propagation of tumor suppressor miRNA Let7b administered with hyaluronic acid-poly(ethyleneimine) (HA-PEI) nanoparticles. Using the experimental data and model fitting, we determined the rate constants for the kinetic events involved in the transfer from the donor cells to the recipient cells. In model 2, we performed Let7b transfection experiments in ID8-VEGF cells with HA-PEI nanoparticles to determine the concentration-effect relationship on HMGA2 mRNA levels. Lastly, in model 3, we combined model 1 and model 2 parameters to describe the kinetics and effect relationship of EV-Let7b in recipient cells to predict the minimum number of miRNA copies needed to show functional effects.


Assuntos
Vesículas Extracelulares , MicroRNAs , Neoplasias Ovarianas , Humanos , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vesículas Extracelulares/metabolismo , Neoplasias Ovarianas/metabolismo , Modelos Teóricos , Microambiente Tumoral
20.
Mol Ther Nucleic Acids ; 30: 64-65, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36213690

RESUMO

The field of gene editing continues to expand significantly and is entering a time of unprecedented utility. Academia and industry look to conquer genetic-based disease with viral and non-viral-delivered CRISPR-Cas9. The most widely used Cas9 protein is derived from Streptococcus pyrogenes (SpCas9), which lends itself to being too large for AAV viral delivery. Cas9 orthologue proteins have diverse size and dependent on bacteria of origin. This diversity has given rise to Cas9 proteins smaller in size while maintaining gene editing abilities. In this article, authors have focused on the use of CjCas9, whose smaller size allows for packaging in AAV and maintains high on-target gene editing. The locus APOC3 was identified for eventual targeting/integration in humans where cardioprotective properties are predicted. To confirm in vivo targeting of this locus, a humanized mouse model was developed due to the absence of the APOC3 locus in mice. These studies looked to answer long-standing questions on integrated gene stability, promoter/low gene integration, and the duration of therapeutic efficacy of the integrated gene.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA