Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Antiviral Res ; 216: 105640, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37263355

RESUMO

Influenza epidemics and pandemics caused by newly emerging virus strains highlight an urgent need to develop a universal vaccine against viruses. Previously, a monoglycosylated X-181mg vaccine demonstrated that the HA possessing a single N-acetylglucosamine at each N-glycosylation site is superior to confer broader protection in mice than conventional vaccines. However, the greatest challenge in conducting clinical trials is the need to develop robust manufacturing processes capable of producing vaccines at the pilot scale with the desired stability, potency, and efficacy. Whether the monoglycosylated virus vaccine platform can be applied to the new vaccine strain in a timely manner and whether the mass-produced vaccine has the proper immunogenicity to induce cross-protective immunity remains unclear. Here, we show that a pilot-scale manufacturing process produced a monoglycosylated A/Brisbane/02/2018(H1N1) virus vaccine (IVR-190mg) with a single glycan at each glycosylation site of HA and NA. Compared with the fully glycosylated virus vaccine (IVR-190fg), the IVR-190mg provided broader cross-protection in mice against a wide range of H1N1 variants. The enhanced antibody responses induced by IVR-190mg immunization include higher hemagglutination-inhibition titers, higher neutralization activity, more anti-HA head domain, more anti-HA stem antibodies, higher neuraminidase activity inhibition titers, and notably, higher antibody-dependent cellular cytotoxicity. Additionally, the IVR-190mg also induced a more balanced Th1/Th2 response and elicited broader splenic CD4+ and CD8+ T-cell responses than IVR-190fg. This study demonstrated that IVR-190mg produced using a pilot-scale manufacturing process elicits comprehensive cross-strain immune responses that have great potential to substantially mitigate the need for yearly reformulation of strain-specific inactivated vaccines.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Camundongos , Humanos , Anticorpos Antivirais , Vacinas de Produtos Inativados , Glicoproteínas de Hemaglutininação de Vírus da Influenza
2.
Biomaterials ; 276: 120989, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34252799

RESUMO

The development of drug delivery strategies for efficacious therapeutic administration directly into the central nervous system (CNS) in a minimally invasive manner remains a major obstacle hindering the clinical translation of biological disease-modifying therapeutics. A novel direct trans-nasal delivery method, termed 'Minimally-Invasive Nasal Depot' (MIND), has proved to be successful in providing high CNS uptake and brain distribution of blood-brain barrier (BBB) impermeant therapeutics via direct administration to the olfactory submucosal space in a rodent model. The present study describes the engineering of custom-made implants with a unique architecture of an "osmotically-active core" entrapping the therapeutic and a "biodegradable polymeric shell" to enable long-acting delivery using the MIND procedure. The MIND-administered implant provided sustained CNS delivery of brain derived neurotrophic factor (BDNF) AntagoNATs for up to 4 weeks in Sprague Dawley rats resulting in significant endogenous BDNF protein upregulation in several brain tissues. The biocompatibility of such core-shell implants coupled with their substantial pharmacokinetic advantages and safety of the MIND procedure highlights the practical utility and translational potential of this synergistic approach for treatment of chronic age-related neurodegenerative diseases.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Sistema Nervoso Central , Sistemas de Liberação de Medicamentos , Animais , Barreira Hematoencefálica , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Mucosa Nasal , Ratos , Ratos Sprague-Dawley
3.
Front Mol Biosci ; 8: 643681, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996898

RESUMO

The possibility of rational design and the resulting faster and more cost-efficient development cycles of nucleic acid-based therapeutics (NBTs), such as antisense oligonucleotides, siRNAs, and gene therapy vectors, have fueled increased activity in developing therapies for orphan diseases. Despite the difficulty of delivering NBTs beyond the blood-brain barrier, neurological diseases are significantly represented among the first targets for NBTs. As orphan disease NBTs are now entering the clinical stage, substantial efforts are required to develop the scientific background and infrastructure for NBT design and mechanistic studies, genetic testing, understanding natural history of orphan disorders, data sharing, NBT manufacturing, and regulatory support. The outcomes of these efforts will also benefit patients with "common" diseases by improving diagnostics, developing the widely applicable NBT technology platforms, and promoting deeper understanding of biological mechanisms that underlie disease pathogenesis. Furthermore, with successes in genetic research, a growing proportion of "common" disease cases can now be attributed to mutations in particular genes, essentially extending the orphan disease field. Together, the developments occurring in orphan diseases are building the foundation for the future of personalized medicine. In this review, we will focus on recent achievements in developing therapies for orphan neurological disorders.

4.
Front Pharmacol ; 12: 660841, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33953687

RESUMO

The most significant obstacle in the treatment of neurological disorders is the blood-brain barrier (BBB), which prevents 98% of all potential neuropharmaceuticals from reaching the central nervous system (CNS). Brain derived neurotrophic factor (BDNF) is one of the most intensely studied targets in Parkinson's disease (PD) as it can reverse disease progression. BDNF AntagoNAT's (ATs) are synthetic oligonucleotide-like compounds capable of upregulating endogenous BDNF expression. Despite the significant promise of BDNF AT therapies for PD, they cannot cross the blood-brain barrier (BBB). Our group has developed an innovative endonasal heterotopic mucosal grafting technique to provide a permanent method of permeabilizing the BBB. This method is based on established endoscopic surgical procedures currently used in routine clinical practice. Our overall goal for the study was to investigate the distribution and efficacy of BDNF AT's using an extra-cranial graft model in naïve rats using the innovative heterotopic mucosal engrafting technique. BDNF AT cationic liposomes (ideal size range 200-250 nm) were developed and characterized to enhance the delivery to rat brain. Uptake, distribution and transfection efficiency of BDNF AntagoNAT's in saline and liposomes were evaluated qualitatively (microscopy) and quantitatively (ELISA and AT hybridization assays) in RT4-D6P2T rat schwannoma cells and in naïve rats. In vivo therapeutic efficacy of BDNF AT's encapsulated in liposomes was evaluated in a 6-OHDA toxin model of PD using western blot and tyrosine hydroxylase immunohistochemistry. Using complimentary in vitro and in vivo techniques, our results demonstrate that grafts are capable of delivering therapeutic levels of BDNF ATs in liposomes and saline formulation throughout the brain resulting in significant BDNF upregulation in key end target regions relevant to PD. BDNF AT liposomes resulted in a better distribution in rat brain as compared to saline control. The delivered BDNF AT's encapsulated in liposomes also conferred a neuroprotective effect in a rat 6-OHDA model of PD. As a platform technique, these results further suggest that this approach may be utilized to deliver other BBB impermeant oligonucleotide-based therapeutics thereby opening the door to additional treatment options for CNS disease.

5.
J Control Release ; 331: 176-186, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33484777

RESUMO

The limitations of central nervous system (CNS) drug delivery conferred by the blood-brain barrier (BBB) have been a significant obstacle in the development of large molecule therapeutics for CNS disease. Though significantly safer than direct CNS administration via intrathecal (IT) or intracerebroventricular (ICV) injection, the topical intranasal delivery of CNS therapeutics has failed to become clinically useful due to a variety of practical and physiologic drawbacks leading to high dose variability and poor bioavailability. This study describes the minimally invasive nasal depot (MIND) technique, a novel method of direct trans-nasal CNS drug delivery which overcomes the dosing variability and efficiency challenges of traditional topical trans-nasal, trans-olfactory strategies by delivering the entire therapeutic dose directly to the olfactory submucosal space. We found that the implantation of a depot containing an AntagoNAT (AT) capable of de-repressing brain derived neurotrophic factor (BDNF) expression enabled CNS distribution of ATs with significant and sustained upregulation of BDNF with efficiencies approaching 40% of ICV delivery. As the MIND technique is derived from common outpatient rhinological procedures routinely performed in Ear, Nose and Throat (ENT) clinics, our findings support the significant translational potential of this novel minimally invasive strategy as a reliable therapeutic delivery approach for the treatment of CNS diseases.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Encéfalo , Administração Intranasal , Barreira Hematoencefálica , Sistemas de Liberação de Medicamentos
6.
Expert Opin Drug Discov ; 13(9): 837-849, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30078338

RESUMO

INTRODUCTION: The central dogma of molecular biology, which states that the only role of long RNA transcripts is to convey information from gene to protein, was brought into question in recent years due to discovery of the extensive presence and complex roles of long noncoding RNAs (lncRNAs). Furthermore, lncRNAs were found to be involved in pathogenesis of multiple diseases and thus represent a new class of therapeutic targets. Translational efforts in the lncRNA field have been augmented by progress in optimizing the chemistry and delivery platforms of lncRNA-targeting modalities, including oligonucleotide-based drugs and CRISPR-Cas9. Areas covered: This review covers the current advances in characterizing diversity and biological functions of lncRNA focusing on their therapeutic potential in selected therapeutic areas. Expert opinion: Due to accelerating parallel progress in lncRNA biology and lncRNA-compatible therapeutic modalities, it is likely that lncRNA-dependent mechanisms of pathogenesis will soon be targeted in various disorders, including neurological, psychiatric, cardiovascular, infectious diseases, and cancer. Significant efforts, however, are still required to better understand the biology of both lncRNAs and lncRNA-targeting drugs. Further work is needed in the areas of lncRNA nomenclature, database representation, intra/interfield communication, and education of the community at large.


Assuntos
Descoberta de Drogas/métodos , Biologia Molecular , RNA Longo não Codificante/genética , Animais , Sistemas CRISPR-Cas/genética , Descoberta de Drogas/tendências , Humanos , Oligonucleotídeos/administração & dosagem
7.
J Ocul Pharmacol Ther ; 34(5): 416-425, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29584529

RESUMO

PURPOSE: To achieve a safer alternative to intravitreal injection of corticosteroids, we developed and characterized triamcinolone acetonide-loaded liposomes formulations (TA-LFs) to be used topically for vitreoretinal drug delivery. METHODS: Four different 0.2% TA-LFs (TA-LF1 to TA-LF4) were generated and submitted to physicochemical characterization. Posteriorly, an ex vivo diffusion assay was performed using rabbit corneas as membranes. Finally, concentrations of triamcinolone acetonide (TA) were determined by high-performance liquid chromatography in ocular tissues from New Zealand white rabbits after multiple topical doses of TA-LF2 (6 times per day, 14 days). In addition, toxicity and tolerability of TA-LF2 was evaluated by cell viability assay and eye examination of study animals, respectively. RESULTS: TA-LF2 was the most stable formulation maintaining a stable hidrogenion potential (pH) at 30 and 40°C and even improving encapsulation with higher temperature. TA-LF2 and TA-LF3 presented the best diffusion performance in vitro reaching the highest TA concentrations after 8 h of follow-up. In vivo diffusion and pharmacokinetics analysis showed that concentrations of TA in retina and vitreous reached the highest peak at 12 h after topical administration of TA-LF2 (252.10 ± 90.00 ng/g and 32.6 ± 10.27 ng/g, respectively) and subsequently decline to 24.0 ± 11.72 ng/g and 19.5 ± 13.14 ng/g, respectively, at 14 days of follow-up. Finally, cell viability was unaffected by TA-LF2, and no increase in intraocular pressure nor ocular alterations were observed after topical administration of this formulation in rabbits. CONCLUSION: TA-loaded liposomes, administered topically, can deliver TA in the vitreous cavity and reach the retina efficiently.


Assuntos
Sistemas de Liberação de Medicamentos , Triancinolona Acetonida/farmacocinética , Administração Tópica , Animais , Sobrevivência Celular , Composição de Medicamentos , Humanos , Lipossomos/administração & dosagem , Lipossomos/farmacocinética , Coelhos , Triancinolona Acetonida/administração & dosagem
8.
Neurobiol Dis ; 77: 35-48, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25725421

RESUMO

Dravet syndrome (DS) is characterized by severe infant-onset myoclonic epilepsy along with delayed psychomotor development and heightened premature mortality. A primary monogenic cause is mutation of the SCN1A gene, which encodes the voltage-gated sodium channel subunit Nav1.1. The nature and timing of changes caused by SCN1A mutation in the hippocampal dentate gyrus (DG) network, a core area for gating major excitatory input to hippocampus and a classic epileptogenic zone, are not well known. In particularly, it is still not clear whether the developmental deficit of this epileptogenic neural network temporally matches with the progress of seizure development. Here, we investigated the emerging functional and structural deficits of the DG network in a novel mouse model (Scn1a(E1099X/+)) that mimics the genetic deficit of human DS. Scn1a(E1099X/+) (Het) mice, similarly to human DS patients, exhibited early spontaneous seizures and were more susceptible to hyperthermia-induced seizures starting at postnatal week (PW) 3, with seizures peaking at PW4. During the same period, the Het DG exhibited a greater reduction of Nav1.1-expressing GABAergic neurons compared to other hippocampal areas. Het DG GABAergic neurons showed altered action potential kinetics, reduced excitability, and generated fewer spontaneous inhibitory inputs into DG granule cells. The effect of reduced inhibitory input to DG granule cells was exacerbated by heightened spontaneous excitatory transmission and elevated excitatory release probability in these cells. In addition to electrophysiological deficit, we observed emerging morphological abnormalities of DG granule cells. Het granule cells exhibited progressively reduced dendritic arborization and excessive spines, which coincided with imbalanced network activity and the developmental onset of spontaneous seizures. Taken together, our results establish the existence of significant structural and functional developmental deficits of the DG network and the temporal correlation between emergence of these deficits and the onset of seizures in Het animals. Most importantly, our results uncover the developmental deficits of neural connectivity in Het mice. Such structural abnormalities likely further exacerbate network instability and compromise higher-order cognitive processing later in development, and thus highlight the multifaceted impacts of Scn1a deficiency on neural development.


Assuntos
Giro Denteado/patologia , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Rede Nervosa/patologia , Convulsões/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Giro Denteado/crescimento & desenvolvimento , Modelos Animais de Doenças , Glutamato Descarboxilase/metabolismo , Hipertermia Induzida/efeitos adversos , Técnicas In Vitro , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Neurônios/ultraestrutura , Convulsões/etiologia , Convulsões/genética , Ácido gama-Aminobutírico/metabolismo
9.
Hum Mol Genet ; 23(R1): R54-63, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24838284

RESUMO

Recent years have seen the increasing understanding of the crucial role of RNA in the functioning of the eukaryotic genome. These discoveries, fueled by the achievements of the FANTOM, and later GENCODE and ENCODE consortia, led to the recognition of the important regulatory roles of natural antisense transcripts (NATs) arising from what was previously thought to be 'junk DNA'. Roughly defined as non-coding regulatory RNA transcribed from the opposite strand of a coding gene locus, NATs are proving to be a heterogeneous group with high potential for therapeutic application. Here, we attempt to summarize the rapidly growing knowledge about this important non-coding RNA subclass.


Assuntos
RNA Antissenso/genética , RNA Antissenso/uso terapêutico , Transcrição Gênica , Expressão Gênica , Marcação de Genes , Genoma , Humanos , RNA não Traduzido/genética
10.
Cell Rep ; 6(1): 222-30, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24388749

RESUMO

Apolipoprotein A1 (APOA1) is the major protein component of high-density lipoprotein (HDL) in plasma. We have identified an endogenously expressed long noncoding natural antisense transcript, APOA1-AS, which acts as a negative transcriptional regulator of APOA1 both in vitro and in vivo. Inhibition of APOA1-AS in cultured cells resulted in the increased expression of APOA1 and two neighboring genes in the APO cluster. Chromatin immunoprecipitation (ChIP) analyses of a ∼50 kb chromatin region flanking the APOA1 gene demonstrated that APOA1-AS can modulate distinct histone methylation patterns that mark active and/or inactive gene expression through the recruitment of histone-modifying enzymes. Targeting APOA1-AS with short antisense oligonucleotides also enhanced APOA1 expression in both human and monkey liver cells and induced an increase in hepatic RNA and protein expression in African green monkeys. Furthermore, the results presented here highlight the significant local modulatory effects of long noncoding antisense RNAs and demonstrate the therapeutic potential of manipulating the expression of these transcripts both in vitro and in vivo.


Assuntos
Apolipoproteína A-I/genética , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , RNA Antissenso/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Apolipoproteína A-I/metabolismo , Células Cultivadas , Chlorocebus aethiops , Células Hep G2 , Hepatócitos , Humanos , Metilação , RNA Antissenso/genética , RNA Longo não Codificante/genética , Células Vero
11.
Pharm Pat Anal ; 2(2): 215-29, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24237027

RESUMO

Over the recent decade oligonucleotides have become an important new class of molecules, allowing therapeutic intervention through targets previously thought 'undruggable'. One of the new avenues opened up by oligonucleotide-based drugs was specific gene upregulation, which, historically, has been difficult to achieve using small-molecule drugs. This article will focus on patents covering this important development in the oligonucleotide field and highlight the different mechanisms through which the oligonucleotide-mediated gene upregulation can work, including inhibition of activity of natural antisense transcripts, interaction with promoter binding sites of noncoding regulatory RNAs, blocking of regulatory and/or miRNA binding sites in 3' UTRs, blocking splice inhibitor/enhancer sites or blocking interactions with polycomb repressive complex 2. Understanding the particular mechanism through which an oligonucleotide drug exerts its effects is highly important in drug development, as it determines the design of the drug molecule.


Assuntos
Oligonucleotídeos/uso terapêutico , Animais , Humanos , Patentes como Assunto , RNA não Traduzido , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA