Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 30(9): 3078-3094, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35821637

RESUMO

mRNA vaccines have recently proved to be highly effective against SARS-CoV-2. Key to their success is the lipid-based nanoparticle (LNP), which enables efficient mRNA expression and endows the vaccine with adjuvant properties that drive potent antibody responses. Effective cancer vaccines require long-lived, qualitative CD8 T cell responses instead of antibody responses. Systemic vaccination appears to be the most effective route, but necessitates adaptation of LNP composition to deliver mRNA to antigen-presenting cells. Using a design-of-experiments methodology, we tailored mRNA-LNP compositions to achieve high-magnitude tumor-specific CD8 T cell responses within a single round of optimization. Optimized LNP compositions resulted in enhanced mRNA uptake by multiple splenic immune cell populations. Type I interferon and phagocytes were found to be essential for the T cell response. Surprisingly, we also discovered a yet unidentified role of B cells in stimulating the vaccine-elicited CD8 T cell response. Optimized LNPs displayed a similar, spleen-centered biodistribution profile in non-human primates and did not trigger histopathological changes in liver and spleen, warranting their further assessment in clinical studies. Taken together, our study clarifies the relationship between nanoparticle composition and their T cell stimulatory capacity and provides novel insights into the underlying mechanisms of effective mRNA-LNP-based antitumor immunotherapy.


Assuntos
COVID-19 , Vacinas Anticâncer , Nanopartículas , Animais , Imunização/métodos , Imunoterapia , RNA Mensageiro/metabolismo , SARS-CoV-2/genética , Baço , Distribuição Tecidual , Vacinação/métodos
2.
Anal Bioanal Chem ; 413(23): 5743-5753, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34476523

RESUMO

Antibody titer and viable cell density (VCD) are two important parameters that need to be closely monitored during the process of cell line development and manufacturing of therapeutic antibodies. Typically, determination of each parameter requires 10-100 µL of supernatant sample, which is not suitable for small scale cultivation. In this study, we demonstrated that as low as 2 µL of culture supernatants were sufficient for the analysis using UV-Vis spectrum assisted with partial least squares (PLS) model. The results indicated that the optimal PLS models could be used to predict antibody titer and VCD with the linear relationship between reference values and predicted values at R2 values ranging from 0.8 to > 0.9 in supernatant samples obtained from four different single clones and in polyclones that were cultured in various selection stringencies. Then, the percentage of cell viability and productivity were predicted from a set of samples of polyclones. The results indicated that while all predicted % cell viability were very similar to the actual value at RSEP value of 6.7 and R2 of 0.8908, the predicted productivity from 14 of 18 samples were closed to the reference measurements at RSEP value of 22.4 and R2 of 0.8522. These results indicated that UV-Vis combined with PLS has potential to be used for monitoring antibody titer, VCD, and % cell viability for both online and off-line therapeutic production process. The process of multivariate analysis and partial least squares regression of UV-Vis spectrum for the determination of CHO cell densities and antibody titers obtained from small volume of cell culture supernatant samples.


Assuntos
Anticorpos/metabolismo , Espectrofotometria Ultravioleta/métodos , Animais , Células CHO , Cricetinae , Cricetulus , Meios de Cultura , Análise dos Mínimos Quadrados
3.
Br J Haematol ; 189(3): 442-451, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31883112

RESUMO

Polyethylene glycol (PEG) conjugated asparaginase (PEGasparaginase) is essential for treatment of paediatric acute lymphoblastic leukaemia. We developed an assay identifying antibodies against the PEG-moiety, the linker and the drug itself in patients experiencing hypersensitivity reactions to PEGasparaginase. Eighteen patients treated according to the DCOG ALL-11 protocol, with a neutralizing hypersensitivity reaction to PEGasparaginase to the first PEGasparaginase doses in induction (12 patients) or during intensification after interruption of several months (6 patients) were included. ELISA was used to measure antibodies, coating with the succinimidyl succinate linker conjugated to BSA, PEGfilgrastim and Escherichia coli asparaginase, and using hydrolysed PEGasparaginase and mPEG5,000 for competition. Anti-PEG antibodies were detected in all patients (IgG 100%; IgM 67%) of whom 39% had anti-PEG antibodies exclusively. Pre-existing anti-PEG antibodies were also detected in patients who not previously received a PEGylated therapeutic (58% IgG; 21% IgM). Antibodies against the SS-linker were predominantly detected during induction (50% IgG; 42% IgM). Anti-asparaginase antibodies were detected in only 11% during induction but 94% during intensification. In conclusion, anti-PEG and anti-SS-linker antibodies predominantly play a role in the immunogenic response to PEGasparaginase during induction. Thus, switching to native E. coli asparaginase would be an option for adequate asparaginase treatment.


Assuntos
Polietilenoglicóis/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Ácido Succínico/uso terapêutico , Feminino , Humanos , Masculino , Polietilenoglicóis/farmacologia , Ácido Succínico/farmacologia
4.
Acc Chem Res ; 52(6): 1555-1564, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31099553

RESUMO

The discovery of CRISPR/Cas has revolutionized the field of genome editing. CRIPSR/Cas components are part of the bacterial immune system and are able to induce double-strand DNA breaks in the genome, which are resolved by endogenous DNA repair mechanisms. The most relevant of these are the error-prone nonhomologous end joining and homology directed repair pathways. The former can lead to gene knockout by introduction of insertions and deletions at the cut site, while the latter can be used for gene correction based on a provided repair template. In this Account, we focus on the delivery aspects of CRISPR/Cas for therapeutic applications in vivo. Safe and effective delivery of the CRISPR/Cas components into the nucleus of affected cells is essential for therapeutic gene editing. These components can be delivered in several formats, such as pDNA, viral vectors, or ribonuclear complexes. In the ideal case, the delivery system should address the current limitations of CRISPR gene editing, which are (1) lack of targeting specific tissues or cells, (2) the inability to enter cells, (3) activation of the immune system, and (4) off-target events. To circumvent most of these problems, initial therapeutic applications of CRISPR/Cas were performed on cells ex vivo via classical methods (e.g., microinjection or electroporation) and novel methods (e.g., TRIAMF and iTOP). Ideal candidates for such methods are, for example, hematopoietic cells, but not all tissue types are suited for ex vivo manipulation. For direct in vivo application, however, delivery systems are needed that can target the CRISPR/Cas components to specific tissues or cells in the human body, without causing immune activation or causing high frequencies of off-target effects. Viral systems have been used as a first resort to transduce cells in vivo. These systems suffer from problems related to packaging constraints, immunogenicity, and longevity of Cas expression, which favors off-target events. Viral vectors are as such not the best choice for direct in vivo delivery of CRISPR/Cas. Synthetic vectors can deliver nucleic acids as well, without the innate disadvantages of viral vectors. They can be classed into lipid, polymeric, and inorganic particles, all of which have been reported in the literature. The advantage of synthetic systems is that they can deliver the CRISPR/Cas system also as a preformed ribonucleoprotein complex. The transient nature of this approach favors low frequencies of off-target events and minimizes the window of immune activation. Moreover, from a pharmaceutical perspective, synthetic delivery systems are much easier to scale up for clinical use compared to viral vectors and can be chemically functionalized with ligands to obtain target cell specificity. The first preclinical results with lipid nanoparticles delivering CRISPR/Cas either as mRNA or ribonucleoproteins are very promising. The goal is translating these CRISPR/Cas therapeutics to a clinical setting as well. Taken together, these current trends seem to favor the use of sgRNA/Cas ribonucleoprotein complexes delivered in vivo by synthetic particles.


Assuntos
Proteína 9 Associada à CRISPR/farmacologia , Sistemas CRISPR-Cas/genética , Portadores de Fármacos/química , Edição de Genes/métodos , Nanopartículas Metálicas/química , Animais , Proteína 9 Associada à CRISPR/genética , Técnicas de Transferência de Genes , Humanos , Camundongos , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/farmacologia , Ribonucleoproteínas/genética , Ribonucleoproteínas/farmacologia
5.
Bioconjug Chem ; 30(2): 461-475, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30188694

RESUMO

Vaccines based on mRNA have emerged as potent systems to elicit CD8+ T cell responses against various cancers and viral infectious diseases. The efficient intracellular delivery of mRNA molecules encoding antigens into the cytosol of antigen-presenting cells (APCs) is still challenging, requiring cell attachment, active uptake, and subsequent endosomal escape. Here, we report a facile approach for the formulation of peptide-functionalized mRNA polyplexes using copper-free click chemistry to promote presentation of mRNA antigen by dendritic cells (DCs). After screening different membrane active peptides, GALA modified mRNA polyplexes (PPx-GALA) with a size around 350 nm and with a slightly negative surface charge (-7 mV), exhibited the highest EGFP-mRNA transfection in RAW 246.7 macrophages (∼36%) and D1 dendritic cells (∼50%) as compared to polyplexes decorated with melittin or LEDE peptides. Interestingly, we found that PPx-GALA enters DCs through sialic acid mediated endo/phagocytosis, which was not influenced by DC maturation. The PPx-GALA formulation exhibited 18-fold higher cellular uptake compared to a lipofectamine mRNA formulation without inducing cytotoxicity. Live cell imaging showed that PPx-GALA that were taken up by endocytosis induced calcein release from endosomes into the cytosol. DCs treated with PPx-GALA containing mRNA encoding for OVA displayed enhanced T cell responses and DC maturation. Collectively, these data provide a strong rationale for further study of this PPx-GALA formulation in vivo as a promising mRNA vaccine platform.


Assuntos
Células Dendríticas/metabolismo , Peptídeos/química , RNA Mensageiro/administração & dosagem , Transfecção/métodos , Animais , Linhagem Celular , Química Click , Proteínas de Fluorescência Verde/genética , Camundongos , Ovalbumina/genética , Polímeros/química , Células RAW 264.7 , RNA Mensageiro/química , RNA Mensageiro/genética
6.
Biomacromolecules ; 20(10): 3641-3647, 2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31418550

RESUMO

Consensus motifs for sequences of both crystallizable and amorphous blocks in silks and natural structural analogues of silks vary widely. To design novel silklike polypeptides, an important question is therefore how the nature of either the crystallizable or the amorphous block affects the self-assembly and resulting physical properties of silklike polypeptides. We address herein the influence of the amorphous block on the self-assembly of a silklike polypeptide that was previously designed to encapsulate single DNA molecules into rod-shaped viruslike particles. The polypeptide has a triblock architecture, with a long N-terminal amorphous block, a crystallizable midblock, and a C-terminal DNA-binding block. We compare the self-assembly behavior of a triblock with a very hydrophilic collagen-like amorphous block (GXaaYaa)132 to that of a triblock with a less hydrophilic elastin-like amorphous block (GSGVP)80. The amorphous blocks have similar lengths and both adopt a random coil structure in solution. Nevertheless, atomic force microscopy revealed significant differences in the self-assembly behavior of the triblocks. If collagen-like amorphous blocks are used, there is a clear distinction between very short polypeptide-only fibrils and much longer fibrils with encapsulated DNA. If elastin-like amorphous blocks are used, DNA is still encapsulated, but the polypeptide-only fibrils are now much longer and their size distribution partially overlaps with that of the encapsulated DNA fibrils. We attribute the difference to the more hydrophilic nature of the collagen-like amorphous block, which more strongly opposes the growth of polypeptide-only fibrils than the elastin-like amorphous blocks. Our work illustrates that differences in the chemical nature of amorphous blocks can strongly influence the self-assembly and hence the functionality of engineered silklike polypeptides.


Assuntos
Capsídeo/química , DNA Viral/química , Peptídeos/química , Multimerização Proteica , Motivos de Aminoácidos , Proteínas do Capsídeo/química , Colágeno/química , Cristalização , Elastina/química , Interações Hidrofóbicas e Hidrofílicas , Seda/química
7.
Mol Pharm ; 14(5): 1482-1493, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28088862

RESUMO

The aim of the present study was to improve the immunogenicity of peptide epitope vaccines using novel nanocarriers based on self-assembling materials. Several studies demonstrated that peptide antigens in nanoparticulate form induce stronger immune responses than their soluble forms. However, several issues such as poor loading and risk of inducing T cell anergy due to premature release of antigenic epitopes have challenged the clinical success of such systems. In the present study, we developed two vaccine delivery systems by appending a self-assembling peptide (Ac-AAVVLLLW-COOH) or a thermosensitive polymer poly(N-isopropylacrylamide (pNIPAm) to the N-terminus of different peptide antigens (OVA250-264, HPV-E743-57) to generate self-assembling peptide epitopes (SAPEs). The obtained results showed that the SAPEs were able to form nanostructures with a diameter from 20 to 200 nm. The SAPEs adjuvanted with CpG induced and expanded antigen-specific CD8+ T cells in mice. Furthermore, tumor-bearing mice vaccinated with SAPEs harboring the HPV E743-57 peptide showed a delayed tumor growth and an increased survival compared to sham-treated mice. In conclusion, self-assembling peptide based systems increase the immunogenicity of peptide epitope vaccines and therefore warrants further development toward clinical use.


Assuntos
Epitopos de Linfócito T/metabolismo , Peptídeos/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/metabolismo , Células Cultivadas , Epitopos de Linfócito T/imunologia , Imunoterapia , Camundongos , Nanopartículas/química , Ovalbumina/química , Vacinação/métodos
8.
Bioconjug Chem ; 27(1): 3-18, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26473310

RESUMO

Self-assembling peptides have gained increasing attention as versatile molecules to generate diverse supramolecular structures with tunable functionality. Because of the possibility to integrate a wide range of functional domains into self-assembling peptides including cell attachment sequences, signaling domains, vaccine epitopes, and even therapeutic moieties, complex nanostructures can be obtained with a wide range of applications in the biomedical field. The first part of this Review provides a concise overview of how peptide primary and secondary structure dictate the way such self-assembling peptides organize into higher ordered, supramolecular structures. Next, an overview of the literature will be given on recent studies on peptide self-assembly for application in drug delivery, vaccination, and tissue engineering.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Peptídeos/química , Engenharia Tecidual/métodos , Vacinação/métodos , Animais , Humanos , Nanoestruturas/química , Peptídeos/metabolismo , Regeneração
9.
J Am Chem Soc ; 137(24): 7775-84, 2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-26022089

RESUMO

Nanovesicles self-assembled from amphiphilic peptides are promising candidates for applications in drug delivery. However, complete high-resolution data on the local and supramolecular organization of such materials has been elusive thus far, which is a substantial obstacle to their rational design. In the absence of precise information, nanovesicles built of amphiphilic "lipid-like" peptides are generally assumed to resemble liposomes that are organized from bilayers of peptides with a tail-to-tail ordering. Using the nanocarrier formed by the amphiphilic self-assembling peptide 2 (SA2 peptide) as an example, we derive the local and global organization of a multimega-Dalton peptide-based nanocarrier at high molecular detail and at close-to physiological conditions. By integrating a multitude of experimental techniques (solid-state NMR, AFM, SLS, DLS, FT-IR, CD) with large- and multiscale MD simulations, we show that SA2 nanocarriers are built of interdigitated antiparallel ß-sheets, which bear little resemblance to phospholipid liposomes. Our atomic level study allows analyzing the vesicle surface structure and dynamics as well as the intermolecular forces between peptides, providing a number of potential leads to improve and tune the biophysical properties of the nanocarrier. The herein presented approach may be of general utility to investigate peptide-based nanomaterials at high-resolution and at physiological conditions.


Assuntos
Nanocápsulas/química , Peptídeos/química , Tensoativos/química , Microscopia de Força Atômica , Simulação de Dinâmica Molecular , Nanocápsulas/ultraestrutura , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , Espectroscopia de Infravermelho com Transformada de Fourier
10.
Mol Pharm ; 12(1): 150-61, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25384057

RESUMO

The applicability of small interfering RNA (siRNA) in future therapies depends on the availability of safe and efficient carrier systems. Ideally, siRNA delivery requires a system that is stable in the circulation but upon specific uptake into target cells can rapidly release its cargo into the cytoplasm. Previously, we evaluated a novel generation of carrier systems ("decationized" polyplexes) for DNA delivery, and it was shown that folate targeted decationized polyplexes had an excellent safety profile and showed intracellular triggered release upon cell specific uptake. Targeted decationized polyplexes consist of a core of disulfide cross-linked poly(hydroxypropyl methacrylamide) (pHPMA) stably entrapping nucleic acids and a shell of poly(ethylene glycol) (PEG) decorated with folate molecules. In the present study, the applicability of folate targeted decationized polyplexes for siRNA delivery was investigated. This required optimization of the carrier system particularly regarding the cross-linking density of the core of the polyplexes. Stable and nanosized siRNA decationized polyplexes were successfully prepared by optimizing the cross-link density of their core. Upon incubation in human plasma, a significant portion of siRNA remained entrapped in the decationized polyplexes as determined by fluorescence correlation spectroscopy (FCS). When tested in a folate receptor overexpressing cell line stably expressing luciferase, Skov3-luc, sequence specific gene silencing was observed. As expected, neither interference on the intrinsic luciferase expression nor on the cell metabolic activity (determined by XTT) was induced by the free-polymer or the siRNA polyplexes. In conclusion, targeted decationized polyplexes are safe and stable carriers that interact with the targeted cells and rapidly disassemble upon cell entry making them promising siRNA delivery systems.


Assuntos
Técnicas de Transferência de Genes , Metacrilatos/química , Nanopartículas/química , RNA Interferente Pequeno/administração & dosagem , Materiais Biocompatíveis/química , Linhagem Celular Tumoral , Ácido Fólico/química , Inativação Gênica , Humanos , Espectroscopia de Ressonância Magnética , Microscopia Eletrônica de Transmissão , Nanotecnologia , Ácidos Nucleicos/química , Polietilenoglicóis/química , Polímeros/química , Espectrofotometria Ultravioleta
11.
Bioconjug Chem ; 25(4): 802-12, 2014 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-24650304

RESUMO

Decationized polyplexes have previously shown unique features, especially regarding their excellent cytocompatibility and very low degree of nonspecific cellular uptake. In the present study, targeted disulfide cross-linked decationized polyplexes were composed of a core of disulfide cross-linked poly(hydroxypropyl methacrylamide) (pHPMA) stably entrapping plasmid DNA (pDNA) and a shell of poly(ethylene glycol) (PEG) decorated with folate molecules. Folate was used as targeting ligand because of its high binding affinity to its receptor, which is overexpressed in many tumors. Studies using folate receptor overexpressing cell lines (HeLa and OVCAR-3) showed significantly higher cell uptake for the folate-targeted decationized polyplexes, when compared to their nontargeted counterparts. On the contrary, for a nonexpressing folate receptor cell line (A549) similar uptake was observed for both targeted and nontargeted decationized polyplexes. Transfection studies using OVCAR-3 cells showed higher transfection efficiency for folate-targeted polyplexes, because of improved cellular uptake. Simultaneously, introduction of targeting moiety on polyplexes did not affect their good cytocompatibilty. The results reported in this paper demonstrate that coupling of folate to decationized polyplexes generates a potential system for targeted gene delivery.


Assuntos
Acrilamidas/química , Sistemas de Liberação de Medicamentos/métodos , Ácido Fólico/metabolismo , Plasmídeos/metabolismo , Polietilenoglicóis/química , Transfecção/métodos , Acrilamidas/metabolismo , Dissulfetos/química , Ácido Fólico/química , Transportadores de Ácido Fólico/genética , Transportadores de Ácido Fólico/metabolismo , Células HeLa , Humanos , Estrutura Molecular , Polietilenoglicóis/metabolismo , Células Tumorais Cultivadas
12.
Microb Cell Fact ; 13: 178, 2014 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-25551787

RESUMO

BACKGROUND: Amphiphilic peptides are important building blocks to generate nanostructured biomaterials for drug delivery and tissue engineering applications. We have shown that the self-assembling peptide SA2 (Ac-AAVVLLLWEE) can be recombinantly produced in E. coli when fused to the small ubiquitin-like modifier (SUMO) protein. Although this system yielded peptides of high purity with no residual amino acids after cleavage of the SUMO fusion protein, the yield after purification was generally low (~1 mg/L bacterial culture) as compared to other peptides and proteins produced with the same method and under the same conditions. RESULTS: The aim of this study is to understand the underlying mechanisms causing the low yield of this recombinant peptide in E. coli and to optimize both production and purification of recombinant SA2 peptides. It was demonstrated that by simply changing the medium to a well-balanced auto-induction medium the yield of recombinant production was augmented (~4 fold). Moreover, it was demonstrated that self-assembly of SUMO-SA2 fusion proteins caused the low peptide yields after purification. By replacing the second IMAC purification step with a selective precipitation step, peptide yields could be increased approx. 3 fold. With these optimizations in place the overall yield of purified SA2 peptide increased with 12-fold. CONCLUSION: Premature self-assembly of the SUMO-SA2 fusion construct interfered with proper purification of the SA2 peptide, resulting in low yields of purified peptide and this could be prevented by changing the mode of purification. These findings are important when setting up purification schemes for other self-assembling peptides with the use of a SUMO fusion construct.


Assuntos
Escherichia coli , Peptídeos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina , Humanos , Peptídeos/química , Peptídeos/genética , Peptídeos/isolamento & purificação , Peptídeos/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/biossíntese , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/isolamento & purificação
13.
Pharm Res ; 31(2): 516-26, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24072262

RESUMO

PURPOSE: Antigen-Expressing Immunostimulatory Liposomes (AnExILs) represent a novel DNA vaccination platform based on the production of protein antigens from DNA templates inside liposomes mediated by an in vitro transcription and translation (IVTT) mix. The aim of this study was to analyze the effects of AnExILs on different dendritic cells (DCs) models and to better understand the role of the different components of this formulation on its adjuvanticity. METHODS: The effect of ß-galactosidase-expressing AnExILs on maturation and particle uptake by murine DC cell line, fresh human monocyte-derived DCs or human dermal DCs in skin explants was investigated and compared to the effects of either plain liposomes or IVTT mix alone. RESULTS: AnExILs induced efficient DC chemotaxis and promoted up-regulation of maturation markers on murine DCs, due to the presence of IVTT in the formulation. Furthermore, the amount of active ßGal associated with DCs was higher for AnExILs than for free ßGal expressed in IVTT or ßGal encapsulated into non-adjuvanted liposomes. Most interestingly, the same trend was observed with human DCs. CONCLUSIONS: Both IVTT mix and liposomal vehicles were shown to be key components of the AnExIL formulation responsible for its adjuvanticity. AnExILs combine antigen production, adjuvanticity and delivery in one system, and can efficiently activate both murine and human DCs.


Assuntos
Antígenos/imunologia , Quimiotaxia/imunologia , Células Dendríticas/imunologia , Lipossomos/imunologia , Pele/imunologia , Animais , Antígenos/genética , Linhagem Celular , Química Farmacêutica/métodos , Quimiotaxia/genética , Células Dendríticas/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Humanos , Imunização , Lipossomos/metabolismo , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Pele/metabolismo , Regulação para Cima/genética , Regulação para Cima/imunologia , beta-Galactosidase/genética , beta-Galactosidase/imunologia
14.
Pharm Res ; 31(11): 3127-35, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24842661

RESUMO

PURPOSE: Since the discovery of RNAi and its therapeutic potential, carrier systems have been developed to deliver small RNAs (particularly siRNA) for modulation of gene expression at the post-transcriptional level. An important factor determining the fate and usability of these systems in vivo is interaction with blood components, blood cells, and the immune system. In this study, a lipid-based and a polymer-based carrier system containing siRNA have been investigated in vitro in terms of their hemocompatibility. METHODS: The nanocomplexes studied were Angiplex, a targeted lipid-based system, and pHPMA-MPPM polyplex, a formulation based on a cationic polymer. siVEGFR-2 was encapsulated in both carriers and activation of platelets, coagulation, and complement cascade as well as induction of platelet aggregation were evaluated in vitro. RESULTS: Both systems had been shown before to cause significant silencing in vitro. Our findings indicated that pHPMA-MPPM polyplex triggered high platelet activation and aggregation although it did not stimulate coagulation substantially. Angiplex, on the other hand, provoked insignificant activation and aggregation of platelets and activated coagulation minimally. Complement system activation by Angiplex was in general low but stronger than pHPMA-MPPM polyplex. CONCLUSIONS: Taken together, these in vitro assays may help the selection of suitable carriers for systemic delivery of siRNA in early preclinical investigations and reduce the use of laboratory animals significantly.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , RNA Interferente Pequeno/química , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Cátions/química , Química Farmacêutica/métodos , Humanos , Lipídeos/química , Metacrilatos/química , Nanopartículas/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Polímeros/química , RNA Interferente Pequeno/administração & dosagem
15.
N Biotechnol ; 83: 56-65, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38945523

RESUMO

Chinese hamster ovary (CHO) cells represent the most preferential host cell system for therapeutic monoclonal antibody (mAb) production. Enhancing mAb production in CHO cells can be achieved by adding chemical compounds that regulate the cell cycle and cell survival pathways. This study investigated the impact of ectoine supplementation on mAb production in CHO cells. The results showed that adding ectoine at a concentration of 100 mM on the 3rd day of cultivation improved mAb production by improving cell viability and extending the culture duration. RNA sequencing analysis revealed differentially expressed genes associated with cell cycle regulation, cell proliferation, and cellular homeostasis, in particular promotion of cell cycle arrest, which was then confirmed by flow cytometry analysis. Ectoine-treated CHO cells exhibited an increase in the number of cells in the G0/G1 phase. In addition, the cell diameter was also increased. These findings support the hypothesis that ectoine enhances mAb production in CHO cells through mechanisms involving cell cycle arrest and cellular homeostasis. Overall, this study highlights the potential of ectoine as a promising supplementation strategy to enhance mAb production not only in CHO cells but also in other cell lines.

16.
Biotechnol Prog ; 40(1): e3403, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37985175

RESUMO

In this study, we investigated the effect of adenosine and its derivative cordycepin on the production yield of a recombinant human monoclonal antibody (adalimumab) in two commonly used Chinese Hamster ovary (CHO) cell lines that have different gene amplification systems, namely CHO-DHFR- and GS-CHO knockout (GS-KO CHO) cells and that were grown in batch culture, with and without glucose feeding. The results showed that adenosine suppressed the cell growth rate and increased the fraction of cells in S phase of the cell cycle for both CHO cell lines. Different concentrations and exposure times of adenosine feeding were tested. The optimal yield of adalimumab production was achieved with the addition of 1 mM adenosine on day 2 after start of the batch culture. Adenosine could significantly improve antibody titers and productivity in both CHO cell lines in cultures without glucose feeding. However, upon glucose feeding, adenosine did not improve antibody titers in CHO-DHFR- cells but extended culture duration and significantly increased antibody titers in GS-KO CHO cells. Therefore, adenosine supplementation might be useful for antibody production in GS-KO CHO cells in medium- to large-scale batches. In case of cordycepin, a derivative of adenosine, CHO-DHFR- cells required higher concentration of cordycepin than GS-KO CHO cells around 10 times to display the changes in cell growth and cell cycle. Moreover, cordycepin could significantly increase antibody titers only in CHO-DHFR- cell cultures without glucose feeding.


Assuntos
Adenosina , Formação de Anticorpos , Desoxiadenosinas , Cricetinae , Animais , Humanos , Células CHO , Cricetulus , Adalimumab , Proteínas Recombinantes/metabolismo , Técnicas de Cultura Celular por Lotes , Glucose/metabolismo
17.
Adv Healthc Mater ; 13(12): e2304238, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38295848

RESUMO

There is no curative treatment for chronic auto-inflammatory diseases including rheumatoid arthritis, and current treatments can induce off-target side effects due to systemic immune suppression. This work has previously shown that dexamethasone-pulsed tolerogenic dendritic cells loaded with the arthritis-specific antigen human proteoglycan can suppress arthritis development in a proteoglycan-induced arthritis mouse model. To circumvent ex vivo dendritic cell culture, and enhance antigen-specific effects, drug delivery vehicles, such as liposomes, provide an interesting approach. Here, this work uses anionic 1,2-distearoyl-sn-glycero-3-phosphoglycerol liposomes with enhanced loading of human proteoglycan-dexamethasone conjugates by cationic lysine tetramer addition. Antigen-pulsed tolerogenic dendritic cells induced by liposomal dexamethasone in vitro enhanced antigen-specific regulatory T cells to a similar extent as dexamethasone-induced tolerogenic dendritic cells. In an inflammatory adoptive transfer model, mice injected with antigen-dexamethasone liposomes have significantly higher antigen-specific type 1 regulatory T cells than mice injected with antigen only. The liposomes significantly inhibit the progression of arthritis compared to controls in preventative and therapeutic proteoglycan-induced arthritis mouse models. This coincides with systemic tolerance induction and an increase in IL10 expression in the paws of mice. In conclusion, a single administration of autoantigen and dexamethasone-loaded liposomes seems to be a promising antigen-specific treatment strategy for arthritis in mice.


Assuntos
Autoantígenos , Células Dendríticas , Dexametasona , Lipossomos , Animais , Lipossomos/química , Dexametasona/química , Dexametasona/farmacologia , Camundongos , Autoantígenos/imunologia , Autoantígenos/química , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Humanos , Artrite Experimental/imunologia , Artrite Experimental/tratamento farmacológico , Artrite Experimental/terapia , Proteoglicanas/química , Proteoglicanas/farmacologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Artrite Reumatoide/imunologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/terapia , Artrite Reumatoide/induzido quimicamente
18.
Eur J Pharm Biopharm ; 196: 114207, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38325664

RESUMO

The discovery that the bacterial defense mechanism, CRISPR-Cas9, can be reprogrammed as a gene editing tool has revolutionized the field of gene editing. CRISPR-Cas9 can introduce a double-strand break at a specific targeted site within the genome. Subsequent intracellular repair mechanisms repair the double strand break that can either lead to gene knock-out (via the non-homologous end-joining pathway) or specific gene correction in the presence of a DNA template via homology-directed repair. With the latter, pathological mutations can be cut out and repaired. Advances are being made to utilize CRISPR-Cas9 in patients by incorporating its components into non-viral delivery vehicles that will protect them from premature degradation and deliver them to the targeted tissues. Herein, CRISPR-Cas9 can be delivered in the form of three different cargos: plasmid DNA, RNA or a ribonucleoprotein complex (RNP). We and others have recently shown that Cas9 RNP can be efficiently formulated in lipid-nanoparticles (LNP) leading to functional delivery in vitro. In this study, we compared LNP encapsulating the mRNA Cas9, sgRNA and HDR template against LNP containing Cas9-RNP and HDR template. Former showed smaller particle sizes, better protection against degrading enzymes and higher gene editing efficiencies on both reporter HEK293T cells and HEPA 1-6 cells in in vitro assays. Both formulations were additionally tested in female Ai9 mice on biodistribution and gene editing efficiency after systemic administration. LNP delivering mRNA Cas9 were retained mainly in the liver, with LNP delivering Cas9-RNPs additionally found in the spleen and lungs. Finally, gene editing in mice could only be concluded for LNP delivering mRNA Cas9 and sgRNA. These LNPs resulted in 60 % gene knock-out in hepatocytes. Delivery of mRNA Cas9 as cargo format was thereby concluded to surpass Cas9-RNP for application of CRISPR-Cas9 for gene editing in vitro and in vivo.


Assuntos
Edição de Genes , Lipossomos , Nanopartículas , Humanos , Feminino , Camundongos , Animais , Edição de Genes/métodos , Sistemas CRISPR-Cas , Proteína 9 Associada à CRISPR/genética , RNA Guia de Sistemas CRISPR-Cas , RNA Mensageiro/genética , Células HEK293 , Distribuição Tecidual , DNA
19.
Int J Pharm ; 655: 124004, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38492899

RESUMO

Photodynamic therapy (PDT) is a suitable alternative to currently employed cancer treatments. However, the hydrophobicity of most photosensitizers (e.g., zinc phthalocyanine (ZnPC)) leads to their aggregation in blood. Moreover, non-specific accumulation in skin and low clearance rate of ZnPC leads to long-lasting skin photosensitization, forcing patients with a short life expectancy to remain indoors. Consequently, the clinical implementation of these photosensitizers is limited. Here, benzyl-poly(ε-caprolactone)-b-poly(ethylene glycol) micelles encapsulating ZnPC (ZnPC-M) were investigated to increase the solubility of ZnPC and its specificity towards cancers cells. Asymmetric flow field-flow fractionation was used to characterize micelles with different ZnPC-to-polymer ratios and their stability in human plasma. The ZnPC-M with the lowest payload (0.2 and 0.4% ZnPC w/w) were the most stable in plasma, exhibiting minimal ZnPC transfer to lipoproteins, and induced the highest phototoxicity in three cancer cell lines. Nanobodies (Nbs) with binding specificity towards hepatocyte growth factor receptor (MET) or epidermal growth factor receptor (EGFR) were conjugated to ZnPC-M to facilitate cell targeting and internalization. MET- and EGFR-targeting micelles enhanced the association and the phototoxicity in cells expressing the target receptor. Altogether, these results indicate that ZnPC-M decorated with Nbs targeting overexpressed proteins on cancer cells may provide a better alternative to currently approved formulations.


Assuntos
Isoindóis , Compostos Organometálicos , Fotoquimioterapia , Humanos , Fármacos Fotossensibilizantes/química , Micelas , Polímeros , Fotoquimioterapia/métodos , Compostos de Zinco , Compostos Organometálicos/farmacologia , Compostos Organometálicos/química , Receptores ErbB , Linhagem Celular Tumoral
20.
Pharmaceutics ; 15(10)2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37896260

RESUMO

The therapeutic potential of the CRISPR-Cas9 gene editing system in treating numerous genetic disorders is immense. To fully realize this potential, it is crucial to achieve safe and efficient delivery of CRISPR-Cas9 components into the nuclei of target cells. In this study, we investigated the applicability of the amphipathic cell-penetrating peptide LAH5, previously employed for DNA delivery, in the intracellular delivery of spCas9:sgRNA ribonucleoprotein (RNP) and the RNP/single-stranded homology-directed repair (HDR) template. Our findings reveal that the LAH5 peptide effectively formed nanocomplexes with both RNP and RNP/HDR cargo, and these nanocomplexes demonstrated successful cellular uptake and cargo delivery. The loading of all RNP/HDR components into LAH5 nanocomplexes was confirmed using an electrophoretic mobility shift assay. Functional screening of various ratios of peptide/RNP nanocomplexes was performed on fluorescent reporter cell lines to assess gene editing and HDR-mediated gene correction. Moreover, targeted gene editing of the CCR5 gene was successfully demonstrated across diverse cell lines. This LAH5-based delivery strategy represents a significant advancement toward the development of therapeutic delivery systems for CRISPR-Cas-based genetic engineering in in vitro and ex vivo applications.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA